Hostname: page-component-6bf8c574d5-qdpjg Total loading time: 0 Render date: 2025-02-23T22:10:01.877Z Has data issue: false hasContentIssue false

FMT and Microbial Medical Products: Generating High-Quality Evidence through Good Governance

Published online by Cambridge University Press:  01 January 2021

Rights & Permissions [Opens in a new window]

Abstract

This article argues that current data for the safety and efficacy of fecal microbiota transplants as a treatment for any indication, including recurrent Clostridioides difficile infection, is low-quality. It develops a governance proposal that encourages production of high-quality evidence by incentivizing well-designed RCTs of stool and stoolderived microbial products. The proposal would require that FDA change its current enforcement approach, but it would not require any change in statutes or regulations.

Type
Symposium 1 Articles
Copyright
Copyright © American Society of Law, Medicine and Ethics 2019

Since data began accumulating from the United States Human Microbiome Project (HMP) and other similar projects around the world,Reference Proctor1 patients, healthcare professionals, and fascinated members of the public have developed tremendous enthusiasm for the possibility that manipulating people's microbial communities can improve their health. The microbial intervention that has garnered the most interest to date is fecal microbiota transplantation (FMT). The theory behind FMT is that microbes in healthy donor stool will repopulate the gut of an ill recipient, restoring microbial diversity in the recipients' gut and providing health-promoting biochemical activity, or inhibiting unhealthy biochemical activity.Reference DeFilipp, Hohmann, Jenq and Chen2 Commentators have proposed FMT as a treatment for a wide range of gastrointestinal and non-gastrointestinal conditions including (but not limited to) inflammatory bowel disease, diabetes, parkinsonism, autism, Alzheimer's disease, graft-versus-host disease in patients with stem cell transplants, depression, and recurrent Clostridioides (formerly Clostridium) difficile infection (rCDI).Reference Lee, Lattimer, Stephen, Borum, Doman, Mileto, Das and Lyras3

Stakeholders' interest in FMT as a medical intervention has been driven by reports that alterations in the composition of people's gut microbial communities are associated with numerous medical conditions;Reference de Vos and de Vos4 by interventional studies in animal models showing that altering the gut microbiota can alter health states, for instance, by inducing or reducing obesity;Reference Bäckhed, Ding, Wang, Hooper, Koh, Nagy, Semenkovich, Gordon, Turnbaugh, Ley, Mahowald, Magrini, Mardis and Gordon5 and by promising results indicating that FMT treats rCDI.Reference Quraishi, Widlak, Bhala, Moore, Price, Sharma and Iqbal6 Strong claims for FMT's effectiveness have led members of the public to engage in do-it-yourself (DIY) FMT, following instructions on websites and YouTube videos.Reference Swaminath, Jeffries and D'Agostino7 DIY FMT has been used to treat a variety of health problems.Reference Ekekezie, Simmons, Perler, Burhke, Duff, Hott, Lillis, Kelly, Subbaraman, Khoruts, Hoffmann and Palumbo8

Despite proponents' enthusiasm, evidence for FMT's efficacy in treating particular medical conditions varies from some to none, is not uniformly high-quality, does not explain how or why the intervention works (if it works), and leaves many unanswered questions about the conditions under which FMT is safe and effective.9 Because stool is widely and cheaply available, healthcare providers and patients have had access to FMT in advance of strong evidence supporting its use.Reference Lietzan10 In addition to DIY FMT, sources estimate that 98% of the U.S. population currently lives within two hours of a healthcare provider who performs FMT.Reference Molteni11

Although cumulative experience with FMT suggests the intervention is relatively low risk, researchers have reported some adverse events,Reference Alang, Kelly, Kelly, Ihunnah, Fischer, De Leon, Watson and Kelly12 including one recent death.13 Several adverse events have been reported following DIY FMT, but this information is anecdotal and no studies have characterized risks of the DIY procedure.Reference Swaminath14

As FMT has become popular, biotechnology companies have begun research and development on microbe-based treatments for rCDI, ulcerative colitis, and other conditions.Reference Williams15 In some cases, companies aim to produce stool-derived microbial products such as defined microbial communities for therapeutic use. This article argues that stool-derived microbial products likely will be at least as safe and effective as stool, and perhaps will perform better.16 Stool-derived microbial products might also compete in the market with stool (used for FMT), and the commercial prospects of stool-derived products can be affected by the way in which stool is regulated (or not regulated).

This article proceeds in four parts. Section I briefly describes the lack of high-quality evidence for stool as a treatment for rCDI or other conditions, suggesting why patients need changes in governance to help incentivize the production of high-quality evidence. Section II compares stool to stool-derived microbial products and argues that stool-derived microbial products likely will be as safe and effective as stool itself, and perhaps better. Regulatory policy should not discourage investment in the development of stool-derived microbial products. Section III discusses how microbiota products for transplantation are regulated and why the FDA's current approach is inadequate for achieving evidence production and other important governance goals. In section IV, the article sets forth a new proposal for governing stool and stool-derived microbial products.

Appropriate governance for stool and stool-derived products should advance several goals and interests. The primary goal of a governance regime should be the production of high-quality evidence regarding the safety and efficacy of microbial products for any indication for which they will be used. Governance should minimize uses of stool or stool-derived microbial products where the risks outweigh the benefits. Governance should also promote, or at least not hinder, medically beneficial innovation, and therefore governance of stool for FMT should not undermine incentives for the development of stool-derived microbial products.

Fortunately, the Food and Drug Administration's (FDA's) authorities to regulate new drugs form a sound basis for a governance regime that can accomplish the above goals. One of the FDA's most important public health functions involves driving the production of high-quality evidence regarding the safety and efficacy of medical products.Reference Eisenberg and Kapczynski17 The FDA's premarket authorities, in particular, provide impetus for private actors to invest in clinical trials. However, applying the new drug regulations to a product already available to patients, such as stool, raises numerous challenges — the access-before-evidence dilemma.18

Prior to 2013, stool for FMT was essentially unregulated and FMT became part of medical practice in advance of high-quality evidence for its safety and efficacy. In 2013, the FDA announced it would regulate stool for FMT as a biological drug, but that it would exercise enforcement discretion regarding the investigational new drug application (IND) requirements for the use of stool for FMT to treat rCDI.19 Some commentators have since argued that enforcement discretion has inhibited the production of high-quality evidence for both stool and stool-derived microbial products as treatments for rCDI, and perhaps, for other indications. This article proposes that the FDA should limit its use of enforcement discretion with regard to stool. Some patients with rCDI who would have received FMT under enforcement discretion ought to be enrolled in clinical studies of FMT or a stool-derived microbial product. Other such patients could receive FMT under the FDA's expanded access program.20

This article proceeds in four parts. Section I briefly describes the lack of high-quality evidence for stool as a treatment for rCDI or other conditions, suggesting why patients need changes in governance to help incentivize the production of high-quality evidence. Section II compares stool to stool-derived microbial products and argues that stool-derived microbial products likely will be as safe and effective as stool itself, and perhaps better. Regulatory policy should not discourage investment in the development of stool-derived microbial products. Section III discusses how microbiota products for transplantation are regulated and why the FDA's current approach is inadequate for achieving evidence production and other important governance goals. In section IV, the article sets forth a new proposal for governing stool and stool-derived microbial products.

Regulation of microbiota transplants is consequential in its own right, given that experts project enormous medical benefit from, and market demand for, such transplants.Reference McQuade, Daniel, Helmink, Wargo, Routy, Gopalakrishnan, Daillère, Zitvogel, Ayyoub, Routy and Kroemer21 However, analysis of policy for microbiota transplants includes broader lessons about the interaction of medical product regulation and innovation policy,22 and should be considered in the context of society's overall commitment to the production of high-quality evidence about all drugs.23

I. Microbiota Transplants Lack High-Quality Evidence

Despite professional and public enthusiasm for FMT as a potential treatment for many ailments, the scientific and medical communities currently lack high-quality evidence of efficacy for the use of FMT to treat any disease.Reference Panchal, Budree and Scheeler24 This article defines “high-quality evidence” as data generated from clinical trials that justifies valid causal inference about the effects of a medical intervention.Reference Friedman, Furberg and DeMets25 This section focuses primarily on evidence for the efficacy of stool for treating medical conditions, particularly rCDI.

Statisticians have long argued that high-quality evidence is best generated through prospective, randomized, controlled, double-blinded, clinical trials.Reference Fisher, Bothwell, Greene, Podolsky and Jones26 Ideally, project personnel conduct such trials according to detailed protocols; they carefully monitor both participants and researchers for adherence to trial protocols; they collect data using detailed case report forms separate from ordinary health records; they use laboratory and clinical tests that elicit information most useful for answering the research question; they rigorously monitor participant outcomes; and they design the study and analyze its results using appropriate statistical expertise, such that the results will most likely lead to valid conclusions.Reference Kendall, Friedman, Furberg and DeMets27 When properly implemented, these features of randomized controlled trials (RCTs) help to produce precise, complete data from which valid conclusions about the safety and efficacy of an investigational medical intervention can be drawn.

Techniques such as randomizationReference Dettori, Bothwell, Greene, Podolsky, Jones and Kendall28 and double-blindingReference Podolsky, Jones, Kaptchuk, Kesselheim, Robertson, Hróbjartsson, Kesselheim, Robertson, Begley and Ioannidis29 help minimize bias and confounding.30 Appropriate controls also help ensure lack of confounding by known prognostic and risk covariates.31 Together, these techniques help researchers make valid causal inferences. They help researchers determine whether a trial's outcomes are caused by the test intervention or by other factors, such as the researchers' or participants' expectations, the natural course of the disease, or some exposure common among people in the treatment arm and not among people in the control arm.Reference Bothwell, Greene, Podolsky and Jones32 Lack of randomization, blinding, and proper controls are reasons why researchers generally cannot use observational studies to determine causality.Reference Sherman, Anderson, Dal Pan, Ejima, Li and Smith33 When the medical community and patients rely primarily on observational and anecdotal evidence, they can engage in medical interventions that do more overall harm than good, or that impose burdens on patients and costs on the medical system without commensurate benefit to patients.Reference Henderson, Paganini-Hill, Ross, Stampfer, Colditz, Grady, Rubin, Petitti, Hulley, Grady and Bush34

When a clinical trial is conducted by drug developers who intend to submit data to the FDA (or a comparable agency) in support of a marketing application, the agency provides oversight of the trial's design, data acquisition, data management and integrity, and data analysis.Reference Lietzan35 Trial sponsors often audit trial sites, and the FDA inspects them. During the review of a sponsor's marketing application, the FDA often conducts an independent analysis of the sponsor's data.36 The FDA expends substantial amounts of money to oversee companies' design and conduct of clinical trials, and then to validate the evidence these trials produce.37 Agency oversight helps clinical trials produce high-quality evidence.38

Enthusiasm for FMT developed in advance of evidence from any double-blinded or well-controlled clinical trial.Reference Kelly, Kim, Laine and Wu39 The strongest evidence for FMT's efficacy is for the treatment of rCDI.Reference Allegretti, Kassam, Osman, Kelly, Khoruts and Staley40 Clostridioides difficile, often a nosocomial infection, causes uncontrollable diarrhea. In 2015, the U.S. Centers for Disease Control and Prevention announced that almost half a million patients were infected with Clostridioides difficile in a single year.41 Of those patients, 29,000 died within 30 days of their initial CDI diagnosis; 15,000 of those deaths were directly attributable to CDI. The number of CDIs is increasing in the U.S. and across the developed world.Reference Lessa, Mu, Bamberg, Edelstein, Daw and Kassam42 Clearly, CDI is an important public health problem in search of an effective treatment.

Arguments for using FMT as a treatment for rCDI were initially based on observational studies, such as case series, and data from small, unblinded trials.Reference Scheeler43 These studies were not designed to produce evidence of efficacy for FDA approval of stool as a treatment for rCDI and were not regulated by the FDA. Results from these initial studies indicated that FMT led to resolution of rCDI symptoms in 89% to 94% of patients.Reference Quraishi, Widlak, Bhala, Gough, Shaikh, Manges, Drekonja, Reich, Gezahegn, Kassam, Lee and Hunt44 Some articles reported a 100% cure rate.Reference Kelly, Kahn and Kashyap45 The first published clinical trial to evaluate the efficacy of FMT for rCDI was a small, unblinded, randomized, active-controlled trail.Reference van Nood, Vrieze and Nieuwdorp46 It was terminated early because FMT appeared quite effective. Taken together, early studies suggested that FMT treated rCDI much more effectively than standard-of-care treatment with antibiotics, and the effect size was so large that many medical professionals may have brushed aside concerns about evidence quality.

Results from the first double-blinded RCT for treatment of rCDI were published in 2016, by which time many patients were clamoring for the intervention and a community of enthusiasts for DIY FMT had already emerged. In the double-blinded study, investigators compared cure rates between participants in the experimental arm, who received donor FMT through colonoscopy, and participants in a control arm, who received autologous FMT.47 “Autologous FMT” means that patient-participants in the control group were administered their own C. difficile-infected feces, prepared the same way as the donor feces were prepared for people in the experimental arm of the study.

The Kelly et al. trial ran at two sites and, unfortunately, results varied substantially between them. Overall, 15 of 24 participants (63%) who received autologous stool were cured (a very high cure rate when compared to antibiotic treatment), whereas 20 out of 22 participants (91%) who received donor stool were cured.48 At one of the two sites, 90% of patients who received autologous stool were cured, as compared to 92% who received donor stool — there was no statistically significant difference between the experimental and control interventions. At the other site, 43% of individuals who received autologous stool were cured, compared to 90% of participants who received donor stool. Across both sites, participants infused with donor stool subsequently showed increased diversity of their gut microbiota. Such diversity is commonly associated with gut health. On the other hand, participants infused with autologous stool continued to display dysbiotic (abnormal) gut micro-biota, with low diversity, even when the participants were cured.

This trial's high between-site variation in cure rates in the control group, and higher-than-expected overall cure rates in the control group, make it difficult to use the data for drawing causal inferences about the effects of donor stool for FMT. The high cure rate for the control group could have occurred by chance. Alternatively, the high cure rate among participants who received autologous stool, and despite their ongoing gut dysbiosis, could indicate that something other than a healthy donor's microbial community cures rCDI when FMT is performed. Based on earlier data, experts have inferred that microbiota in healthy donor stool caused the resolution of symptoms;Reference Seekatz, Rao, Santhosh and Young49 however, that conclusion cannot be drawn from existing data.

The NIH recently funded a placebo-controlled, double-blinded RCT to assess the safety and efficacy of treating rCDI using FMT delivered by enema.50 This trial started in 2018 and is now recruiting participants.51 Whether this public investment will be sufficient to answer important questions concerning the causal relationships between microbes in stool and resolution of rCDI symptoms remains to be seen.

While there is little high-quality evidence that microbes in stool cure rCDI, the anecdotal evidence for FMT as a cure is notable. There are cases in which people on the verge of death from rCDI received FMT and made a full recovery.52 This article's questioning of the evidence base is not meant to denigrate the profound experiences of people who have undergone FMT and been cured, but rather to say there is insufficient evidence that fecal microbiota cure rCDI. Perhaps, a colonoscopy that delivered saline would also have cured some or most of those people. Even if some people are cured by fecal microbiota, without good evidence nobody knows who is likely to benefit from FMT or by which microbes they would be helped.

In 2018 the Infectious Disease Society of America and the Society for Healthcare Epidemiology of America updated their clinical practice guidelines to include a strong recommendation for FMT to treat adults with rCDI.Reference McDonald, Gerding and Johnson53 Some private health insurers now cover this treatment.54 Medicare and its contractors also pay for it.Reference Lessa, Mu and Bamberg55 Due to the absence of conventional medical alternatives, FMT as a treatment for rCDI has become widely available based on low-quality evidence showing strong effects of FMT.

The apparent success of FMT as a treatment for rCDI, and other evidence that dysbiosis of the gut microbiota is associated with many illnesses, have led to widespread enthusiasm for FMT as a possible treatment or cure for a wide variety of ailments other than rCDI.Reference Vrieze, de Groot and Kootte56 In June of 2019 at least 310 clinical studies were investigating FMT for some indication, although only a few of those studies were double-blinded RCTs.57

Despite the value of well-designed RCTs, various trends in science and medicine have decreased public, medical professional, and Congressional enthusiasm for them. These trends include: their high costs; what some stakeholders perceive as their excessively slow pace; the difficulty of generalizing RCT results to broader, real-world patient populations; the difficulty of conducting RCTs for rare or “molecularly subsetted” diseases;58 the difficulty or impossibility of detecting rare but serious side-effects in an RCT; the rise of patient-driven research; and the increasing availability of health care data for use in big data analyses.Reference Sherman, Anderson and Dal Pan59 In response to the preceding concerns and opportunities, many stakeholders hope to use real world evidence (RWE) to supplement or supplant evidence from RCTs. Data for producing RWE include (but are not limited to): patient-reported outcomes, electronic health record information, claims and billing information, data collected from product and disease registries, data from healthcare operations, social media posts or queries, and information gleaned from wearables and apps.60

RWE could be generated for FMT even though stool for FMT has not been approved for any indication, because FMT is occurring in clinical practice and DIY settings. FMT is also being conducted outside of the U.S. for a variety of indications.Reference Edelstein, Kassam and Daw61 A coalition of U.S. medical professional societies and medical philanthropies has created an FMT national registry that includes some data from which RWE could be generated.62 Such evidence could be useful in understanding long-term or low frequency safety issues associated with FMT. However, given the complexity of the gut microbiota and their relationship to health and disease, it seems unlikely that decision-makers, including researchers, clinicians and regulators, could or should use RWE as a basis for valid judgements about FMT's efficacy for treating any indication.

To the extent that RWE derives from observational data, it suffers from the same problems as other observational evidence discussed above — it is not generally suitable for making causal inferences. Even when RWE is generated in a prospective experiment, the data sources for RWE are prone to error, missingness, and numerous biases.Reference Sherman, Anderson, Dal Pan, Pearl, McMurray, Freemantle, Marston and Walters63 Data for RWE typically lack the quality controls imposed in a well-designed and properly-conducted RCT. Analysts may find it impossible to sort out fundamental issues of confounding and bias in such data. Data scientists are working to develop methods for producing high-quality RWE,Reference Berger, Sox and Wilke64 and even for drawing causal inferences from observational data,Reference Kuang, Bao, Thomson and Vanhaelen65 but to date most experts caution that RWE should supplement, rather than supplant, evidence from well-designed RCTs.Reference Gottlieb66 Governance of microbiota for transplantation cannot rely on RWE to fill in evidence gaps left by a dearth of RCTs.

Thus far, this article has focused on evidence for FMT's efficacy; however, a brief discussion of safety is also in order.Reference Ossorio and Zhou67 Experts worry about the risks of transmitting dangerous microorganisms and unhealthy phenotypes (such as obesity or depression), physically injuring recipients when administrating FMT (e.g., by colonoscopy or nasogastric tube), and exacerbating a recipients' disease.Reference Baxter and Colville68 Unfortunately, much existing safety data comes from metanalyses of case studies and case series in which safety information was not systematically collected.69 The data are generally low-quality and relate primarily to short-term risks. Studies in which safety data were systematically collected suggest that FMT administered by a healthcare provider has some risks, but their probability is low and they are generally mild and tolerable.Reference Qazi70

Transmission of pathogenic organisms is a primary concern for experts, patients, and research participants.Reference Paramsothy, Borody, Lin, Park, Mone and Price71 When FMT is conducted by health care providers the donor and stool are screened to avoid transmission of infectious diseases and unhealthy phenotypes.Reference Dubois, Ling and Osman72 There are no widely accepted professional standards for how donors and stool should be screened,Reference Wood-worth, Neish, Miller, Terveer, van Beurden and Goorhuis73 and there is little guidance from the FDA on this issue;74 however, responsible stool providers conduct extensive screening.Reference Dubois, Ling and Osman75 Despite donor and stool screening, the possibility of transmitting pathogens via FMT remains, either because a screening test missed a known pathogen or because a new pathogen has entered the human microbiota and there are no tests to detect it.76

While this article was in production, the FDA announced that two immunocompromised patient-participants in an FMT study had contracted a multi-drug resistant, pathogenic strain of E. coli from FMT. One participant died from the infection.77 The FDA issued instructions for screening donors and stool for multi-drug resistant microorganisms, and required that all researchers licensed to conduct FMT studies (all researchers using FMT under an IND78) implement such screening by July 15, 2019.79

Finally, note that scientists and regulators cannot evaluate the safety of FMT vel non. FMT could have different safety profiles in patients with different diseases. The way in which transplanted microbes engraft in a recipient, and whether or not they cause harm, could depend on how a recipient's gut has been altered or damaged by her underlying disease.

This section has presented the case that the evidence base for FMT as a safe and effective treatment is weak. Past governance approaches have not incentivized the production of high-quality evidence. The next section will compare FMT to stool-derived microbial products, and argue that stool-derived microbial products will likely be as good or better than stool for treating disease.

II. Stool Versus Stool-derived Microbial Products

Stool is a complex mixture of bacteria, viruses, fungi, human cells, mucous, extracellular proteins, and metabolites.Reference Hoffmann, Palumbo and Ravel80 Stool for FMT can be delivered via colonoscopy, nasogastric tube, enema, or capsules.81 Stool can be prepared and screened “in-house” by clinicians and pharmacists at a health clinic or hospital, or it can be produced and shipped by a stool bank.82 For purposes of this article, stool banks are entities that obtain, prepare, and store stool for later distribution to other institutions for use in patient therapy or clinical research.83

One problem in developing an evidence base for the efficacy and safety of FMT is that stool has not been well-characterized. Over half a decade ago, work from the HMP suggested that human health and disease might not be determined by the taxa of microbes in people's microbial communities, but rather by the collection of molecular functions performed by the specific strains in those communities.84 Yet, a recent commentary notes that researchers have done little to determine which changes in microbiota composition or function represent causes of disease, rather than effects or associations.Reference Lloréns-Rico and Raes85 If microbes in stool produce beneficial effects through FMT, researchers still do not know which microbes do so, or why.

Complexity does not make stool unusual among items regulated as biological products, which the FDA describes as “complex mixtures that are not easily identified or characterized.”86 Stool is, however, orders of magnitude more complex than many other biological products. Because most biological products cannot be fully characterized, regulators define them in part by their manufacturing processes, which are tightly controlled.87 The FDA's oversight of stool manufacturing focuses on the processes for donor and stool screening, methods for stool storage and preparation, effects of adding saline or stabilizers, techniques for characterizing stool, and other relevant considerations.Reference Slater88

However, the FDA has no jurisdiction over the processes by which stool is initially produced in and excreted from the human body. Stool is difficult to characterize not only because it is complex, but because it is dynamic and responsive to the environment.89 The composition of a donor's stool will vary with the person's diet, general health, behaviors (such as the amount of sleep the person had before donating), perhaps time of day, and other unknown factors. Characterizing stool from every donor at every donation, for instance by developing a genomic profile of all microorganisms the stool contains, would be impossible or prohibitively expensive using current technology.90

Because stool for FMT cannot be well-characterized, it is all the more important for researchers, regulators, and clinicians to understand which, if any, of its components have therapeutic effects. If scientists gain a better understanding of which microbes, and which microbial functions or molecules, cause changes in a person's health, then clinicians and researchers should be able to adequately characterize stool: they should be able to measure the components that matter when FMT is used as a treatment. If researchers identify which microorganisms and microbial functions cause health improvements, they can develop metrics for stool's potency and stability in relation to different methods of storage, preparation, and delivery. Currently, these basic characteristics of a biological drug are not determinable for stool.

Users cannot determine what dose of a therapeutic agent is being administered in a specified quantity of stool. With microbiota the starting dose might not be as important as it is for a small molecule drug; after all, microbes can multiply. However, without knowledge of which microorganisms or molecules are therapeutic for a given indication, researchers, regulators, and clinicians cannot know whether any therapeutic agent has been delivered to a recipient or whether one donor's stool will be more therapeutic than another donor's stool for a particular indication. They cannot know whether the therapeutic agent has grafted at its target body site, whether it has multiplied, or whether it is producing therapeutic molecules or functions in the new human host. In addition, the scientific community has little understanding of which genetic, metabolic, or other “ecological elements” of an FMT recipient influence the activity or treatment effects of donor microbiota.91

Bacteria are the easiest microorganisms to identify and characterize in the gut microbiota, but they are not the only gut microorganisms influencing human health. Fungi and viruses are also present and may influence the course of many diseases.Reference Sokol, Leducq, Aschard, Chehoud, Dryga, Hwang, Lloyd-Price, Arze and Ananthakrishnan92 Adequately characterizing the gut microbiota, and their transformations, will involve a better understanding of viruses and fungi.

Private firms face disincentives to developing stool as an approved drug. As a product of nature, minimally-manipulated stool will be difficult to patent.93 Patent protection will be available for new and non-obvious methods of storing, freezing, or administering stool, and for products containing stool in specialized capsules or other delivery vehicles.Reference Stevens, Sandowsky, Khoruts, Weingarden and Hamilton94 Thus far, such patents have not incentivized substantial private investment in developing stool as a drug to treat rCDI or any other indication, perhaps because stool is widely available outside of firms' control, the types of patents available are easy for people to work around, or patent holders might have difficulty enforcing their patents against large numbers of small-volume infringers. A combination of public funding, philanthropic, and non-profit investments could lead to development of stool as a drug, but thus far has led to numerous small studies and a few RCTs that have not produced the quality of data that generally would lead to marketing approval.

Given clinicians' and patients' enthusiasm for microbiota transplants as a treatment, and also the difficulties and risks of using stool as a drug, it should be no surprise that biotechnology companies are attempting to develop stool-derived microbial products for treating rCDI and other medical conditions.Reference Gertner95 For purposes of this article, “stool-derived microbial products” consist of defined communities of microbes that were originally isolated from stool samples. Such communities could range from complex consortia to relatively simple mixtures.96 They will be composed of known strains, and to create such a product scientists must identify which strains have therapeutic properties. Compared to stool, a stool-derived microbial product will contain few or no human cells and proteins, and far fewer taxa of bacteria, viruses, and fungi.

Stool-derived microbial products will raise some of the same regulatory challenges as stool and are likely to compete in the same markets as stool.97 These products are regulated as biological drugs and do not receive any enforcement discretion for their use. No such product is currently on the market, although several are in development.

As drugs, stool-derived microbial products will likely be as good as or better than stool.98 Once such defined mixtures or consortia of microbes are created, specifying and characterizing their important functionalities, and standardizing the microbial product, should be more straightforward than doing so for stool. Knowledge of which microbes and molecules provide therapeutic benefit will facilitate assessment of the product's purity, potency, dosing, stability and other important characteristics related to delivering safe and effective drugs to patients. Stool-derived microbial products might also have fewer off-target effects than stool, because stool-derived products will contain fewer microbes and other biologically active materials.

As a practical matter, stool-derived microbial products likely will be grown in laboratories rather than repeatedly isolated from stool. Laboratory-grown products would be less likely than stool to acquire human gut pathogens. And because even a complex microbial consortium would be less complex and far better characterized than stool, a stool-derived microbial product would be easier to screen for contamination by microorganisms not intended as part of the product.

For the above reasons, stool-derived microbial products likely will be safer and as or more effective drugs than stool for many indications for which microbes cause resolution of symptoms. FMT using stool from human donors might best be viewed as a temporary solution to treating medical problems, and used only until a stool-derived microbial product or other safe and effective drug becomes available.99

In light of the scientific community's current lack of knowledge regarding the therapeutic functionalities of microbes in stool, biotechnology companies are undertaking extensive preclinical and clinical research to develop stool-derived microbial products. Such research takes time and costs significant sums of money, and when stool-derived microbial products come on the market they will likely be more expensive than well-screened stool.100 Whether clinicians and patients find the additional costs justifiable will depend on the price differentiation, the relative effectiveness of the stool-derived microbial products, their ease of administration, and their marketing.

Whether firms continue to invest in development of stool-derived products will depend, in part, on how stool for FMT is regulated. Expected future returns shape firms' incentives to make present investments in research and development of new drugs (including biologics), and regulation can influence a firm's expected returns.Reference DiMasi, Grabowski, Danzon and Nicholson101 If the FDA continues its current enforcement discretion policy, and particularly if the FDA signals that it might leave unapproved stool on the market after a stool-derived microbial product is approved, biotechnology companies will have a dis-incentive to invest in stool-derived products. If unap-proved stool, sold by stool banks that did not need to recoup the costs of RCTs and FDA approvals, could compete in the same market as an approved stool-derived microbial product, then rational firms would be less likely to invest in producing a stool-derived product.102

III. Enforcing Drug Regulations for Stool and Stool-Derived Products: The FDA's Current Approach

Since 2013, the FDA has regulated microbiota for medical transplantation as a biological drug, whether the microbiota consist of stool or a stool-derived microbial product.103 A new drug, including a new biological drug, must undergo premarket review in which the sponsor presents the FDA with substantial evidence for the drug's safety and efficacy.104 Substantial evidence has traditionally meant evidence from two prospective, double-blinded RCTs. However, that quantity and quality of evidence is not required by law, and the FDA has flexibility regarding the amount and type of evidence it accepts.105

For a biological drug such as stool, a Biologics License Application (BLA) must be approved prior to the drug entering the market.106 Approval of a BLA, and the twelve years of data exclusivity that accompanies such approval, are mechanisms by which FDA oversight incentivizes firms to invest the hundreds of millions, perhaps billions, of dollars required to develop high-quality evidence of a biological drug's safety and efficacy.107 Firms sometimes game the evidence-production system, for instance, by designing clinical trials to avoid learning important information,108 but no other set of institutional arrangements and incentives has produced as much or as good medical evidence as the FDA's authorities to regulate new drugs.

Prior to approval of a BLA, researchers must obtain an investigational new drug application (IND) before using the drug in clinical research.109 Outside of research, manufacturers can only provide an unap-proved drug to clinicians and patients under a narrow set of circumstances; although, the U.S. market for pharmaceuticals includes numerous unapproved drugs that are sold openly but illegally.110 Clinicians and patients may attempt to legally obtain an unap-proved drug using the FDA's expanded access program (often referred to as “compassionate use”),111 or using a pathway created by the new federal “Right to Try” law.Reference Wendler, Mongiello, McLinn and Bellina112 Both of these pathways restrict the kinds of patients who are eligible to receive a drug and the types of drugs that can be provided, and neither pathway requires that a manufacturer provide the unap-proved drug.

As mentioned above, prior to 2013 stool for FMT was not regulated and FMT was incorporated into medical practice as a treatment for rCDI. Now, facing the access-before-evidence dilemma, the FDA is struggling to regulate stool. In May of 2013, “everyone kind of flipped out”Reference Fischer, Kassam, Kelly and Sobcinski113 when the agency announced that it would regulate FMT as a biological drug. Later that year, the agency published guidance announcing that it would exercise enforcement discretion for microbiota used to treat “C. difficile infection not responding to standard therapies.”Reference Price and Price114 This policy permits manufactures to produce and distribute stool without an IND when the stool will be used to treat rCDI. Distribution of stool for all other indications still requires an IND.

Under the 2013 guidance, stool banks have shipped over 40,000 stool preparations to clinicians for use in FMT,115 and clinicians have conducted an unknown number of FMTs using in-house-produced stool. While the guidance only extends enforcement discretion to stool for treating rCDI, the FDA will not always know whether clinicians are using stool to treat other indications, particularly if that stool was produced in-house. One biotechnology firm stated that some entities “are investigating and marketing FMT products for other indications [not rCDI] without complying with the applicable regulatory requirements.”116

Numerous commentators have argued that the FDA should not regulate stool for FMT as a drug, or that the agency should not to regulate stool at all.Reference Hoffmann, Palumbo, Ravel, Smith, Kelly, Alm, Sachs, Edelstein, Riley and Olle117 Such arguments entail a claim that stool should not be required to undergo the type of clinical trials and regulatory scrutiny that could produce high-quality evidence of its safety and efficacy for treating particular medical conditions. Such a claim might be credible if high-quality evidence for medical use of FMT, even to treat rCDI, had been produced during the pre-2013 period in which stool was unregulated. However, in the absence of FDA drug regulation there were and are no incentives strong enough to promote investments of the magnitude needed to produce high-quality evidence for FMT.118

In 2014 and 2016 the FDA attempted to update its 2013 policy regarding enforcement discretion for the use of stool to treat rCDI.119 The agency published draft guidance announcing that it would not extend enforcement discretion to stool banks. It proposed that a stool bank should either hold an IND or ship product only to an entity with an active IND. The agency raised concerns that centralized manufacturing and distribution of stool could create significant public health risks.120 If a stool bank failed to adequately screen donors or stool, and distributed a dangerous product to numerous different hospitals for use in numerous patients, a great deal of harm could occur. Under enforcement discretion, the FDA would not know which stool banks were distributing products to which medical providers. It would not know how the donors and stool were screened. The agency would not receive systematically collected adverse event reports. Under such circumstances, the FDA and the medical community would be slow to detect safety problems. The 2014 and 2016 draft guidances likely were influenced by the specter of a public health fiasco that occurred in 2012, in which a single compounding pharmacy distributed contaminated drugs across the U.S., causing fungal meningitis in 753 people and killing sixty-four.Reference Outterson121

Many commentators voiced strong opposition to both the 2014 and the 2016 proposals. Some medical experts argued that the proposed policies could move the preparation and screening of stool from stool banks to small healthcare organizations that lack the scientific expertise, logistical capacity, and financial wherewithal to conduct the extensive screening that stool banks currently perform.122 Such an outcome would make FMT more dangerous, undermining the FDA's public health objectives. Other commentators were concerned that requiring an IND to use materials from stool banks would make FMT for rCDI less available to patients, because clinicians with the expertise to administer FMT might lack the time or expertise to file IND paperwork. After considering such criticism, the FDA did not finalize either of the draft guidance documents.

Operating under the 2013 guidance, stool banks still distribute material for FMT to physicians, without an IND, for use in treating rCDI.Reference Edelstein123 The first and best-known stool bank in the U.S., the non-profit OpenBiome, also helps clinicians to obtain INDs so the bank can distribute stool to them for uses other than treating rCDI.124 OpenBiome has a “biologics master file” with the FDA,125 and clinician-investigators not employed by the stool bank can reference this file when applying for their own INDs.

Some microbiome scientists have argued that the FDA's enforcement discretion policy has slowed evidence generation for stool and for stool-derived microbial products.126 One expert lamented that “only a tiny fraction of data on the >10,000 patients treated by OpenBiome products has been systematically collected and published.”Reference Khoruts127 In addition, several commentators believe that widespread access to FMT has slowed enrollment in RCTs testing both stool and stool-derived products as treatments for rCDI. Potential participants may be less likely to enter a trial in which they might be randomized to a placebo or other control arm;Reference Kelly, Fischer, Grinspan and Allegretti128 they might not receive FMT immediately; or they might receive a stool-derived microbial product instead of donor stool.129 When exercising enforcement discretion, the FDA has no opportunity to assess whether access to the unapproved product is hindering the evidence generation process.

Another reason some patients might not be enrolling in RCTs testing potential treatments for rCDI is because their clinicians believe that the medical community is no longer in equipoise about the value of FMT. Based on recently promulgated practice guidelines recommending FMT for treatment of rCDI,130 and insurers' increasing willingness to pay for the intervention, clinicians might believe it is unethical to recommend that their patients participate in a trial if they could be randomized to an arm in which they do not receive FMT. Note that both physician and patient concerns about participation in RCTs are likely premised on the belief that microbiota transplants are safe and effective treatments for rCDI or other indications. This article argues that belief is not yet well-justified, even for rCDI.

IV. A Proposal for Governing FMT and Stool-derived Microbial Products

Enforcement discretion should not be a permanent solution to the access-before-evidence dilemma with regard to FMT for rCDI (or for other potential drugs). Access-before-evidence leaves society with the continuous accumulation of low-quality and potentially misleading evidence, while inhibiting the production of high-quality evidence for the use of stool or stool-derived microbial products. Enforcement discretion may decrease incentives for private investment in the development of stool-derived products, even though such products would likely be safer and as or more effective than stool. The FDA would better fulfill its public health mission by curtailing its use of enforcement discretion for stool. Both the 2014 and 2016 draft guidance documents indicate the agency's intention to end enforcement discretion in regard to stool distributed by banks.

The policy articulated in the 2016 draft guidance is practicable, and is superior to the current enforcement discretion policy; however, this article proposes a slightly different approach:

  • Enforcement discretion should not extend to any use of stool by institutions where any investigator is currently conducting research under an IND, or where any investigator has done so in the recent past (probably, the last three years).

  • Stool to treat rCDI meets the criteria for expanded access for an intermediate-size patient population.131 When appropriate, the FDA should grant expanded access to stool for FMT to treat intermediate-size patient populations with rCDI.132 The agency should prioritize expanded access for organizations that (1) sponsor an IND for treatment of rCDI with stool or a stool-derived microbial product; (2) employ at least one investigator133 who, under an IND, is conducting studies of stool or a stool-derived microbial product to treat rCDI; or (3) can demonstrate that they have successfully referred at least one patient to a relevant RCT.

    • – Consistent with the regulations, the FDA should only grant expanded access if “providing the investigational drug for the requested use will not interfere with the initiation, conduct, or completion of clinical investigations that could support marketing approval of the expanded access use or otherwise compromise the potential development of the expanded access use.”134

  • The FDA should undertake extensive and varied stakeholder engagement activities, with the aim of developing RCT designs for FMT and stool-derived microbial products that are (1) attractive to patients and their clinicians; (2) capable of efficiently producing data the FDA will accept in support of a BLA; and (3) feasible and affordable.

This proposal for changing regulatory enforcement aims to promote the generation of high-quality evidence for the use of stool in FMT, and for stool-derived microbial products. It would require that clinicians at organizations with significant research and regulatory sophistication obtain an IND before undertaking FMT for any reason, including treatment of rCDI. They would need an IND for using any stool, whether from a bank or produced in-house. Although obtaining an IND is burdensome, the proposal would impose these burdens on investigators at “research-capable” institutions, which already possess the infrastructure to participate in FDA-regulated research and are capable of producing high-quality evidence.

This proposal for changing regulatory enforcement aims to promote the generation of high-quality evidence for the use of stool in FMT, and for stool-derived microbial products. It would require that clinicians at organizations with significant research and regulatory sophistication obtain an IND before undertaking FMT for any reason, including treatment of rCDI.

Obtaining an IND is feasible for research-capable institutions and their scientists. Sponsor-investigators135 at some research-capable healthcare institutions already hold research-INDs for use of stool to treat people with rCDI or other conditions,Reference Kelly, Kunde and Kho-ruts136 and many hold INDs for studying other drugs. The medical literature instructs clinician-scientists in how to obtain a research-IND;Reference Holbien137 the FDA provides instructions;138 and some academic centers have offices that help clinician-scientists apply for INDs. Just as Open-Biome currently does for non-rCDI indications, stool banks should have a biologics master file and permit sponsors to reference that file when submitting their INDs.

When the product under study is a stool-derived microbial product, the IND likely will be held by a commercial sponsor and researchers at the healthcare institution will be investigators139 (not sponsor-investigators). Healthcare institutions whose investigators conduct research under a commercial sponsor's IND are selected by the sponsor because of the institutions' infrastructure for conducting regulated research and its researchers' superior reputations. Such institutions should be producing high-quality evidence for the use of FMT and stool-derived products.

Under this article's proposal, healthcare providers and institutions with less research sophistication — often smaller, non-academic, less-well-resourced, or in less populous areas — would not need an IND for obtaining materials from a stool bank or for preparing stool in-house. Most of these institutions probably see few patients with rCDI. If they provide FMT, they may lack the resources to safely prepare and store stool in-house, so policies should permit and even encourage them to obtain FMT materials from a stool bank. Continued enforcement discretion for them likely will not undermine the generation of high-quality evidence, but it will promote patient safety.

Requiring that certain providers of FMT have an IND is not the same as requiring that they all conduct RCTs. The IND regulations only specify that the drug be used in a “clinical investigation,” which is defined as “any experiment that involves a test article and one or more human subjects…”140 Clearly, some patients who seek FMT for treatment of rCDI would be ineligible or unable to participate in an RCT. Some rCDI patients might not have an RCT geographically accessible. Some rCDI patients would not meet eligibility criteria for an RCT. For instance, a recent study found that 75% of patients with rCDI were not eligible to participate in an RCT testing a stool-derived product.141 The problem of restrictive eligibility criteria that slow accrual to RCTs is not unique to studies of FMT;Reference Kim, Atlas, Ison and Ersek142 the FDA and other stake-holders are working to address this issue. In addition to requiring that sponsors justify eligibility criteria for trials of FMT or stool-derived products to treat rCDI, the FDA can work with sponsors to design some non-RCT studies for which some people who cannot participate in RCTs would be eligible. This approach will not always produce the highest quality data, but it ought to produce some data and systematic adverse event reporting, which is more than society receives under the current enforcement discretion policy.

Although the regulations permit INDs for studies that are not RCTs, this article's proposal aims to encourage faster accrual for RCTs. It does so by using the FDA's expanded access authorities rather than enforcement discretion. When considering whether expanded access would “interfere with the initiation, conduct, or completion of clinical investigations,”143 the FDA can and should consider whether it is receiving enough INDs for RCTs for stool and stool-derived products, and whether existing RCTs are accruing participants. Further, the regulation for expanded access for intermediate-size patient populations states that if a relevant clinical trial is underway, sponsors of the expanded access IND/protocol must “explain why the patients to be treated cannot be enrolled in the clinical trial and under what circumstances the sponsor would conduct a clinical trial in these patients.”144 The FDA probably has authority to require that patients be referred to a relevant RCT (one that is currently accruing participants) before they are considered for treatment under expanded access. This article's proposal would also incentivize clinicians at research-capable institutions to refer patients to RCTs by prioritizing the grant of expanded access INDs to entities that successfully refer patients to RCTs.

The proposal would decrease patients' access to FMT outside of studies, at least for patients who receive care at research-capable institutions. Given the goal of producing better evidence, incentivizing patients to receive FMT or a stool-derived microbial product through a study is a feature of the proposal. Patients who are ineligible or unable to participate in a study often would be eligible for expanded access. If a patient did not qualify for expanded access, there might be a good reason why she or he should not be receiving FMT. Overall, the aim of this proposal is not to substantially change the number of people who receive unapproved stool for FMT, but to change the context in which they receive it to one in which more and better evidence can be generated.

Expanded access requires submissions to the FDA, and therefore places burdens on clinicians and their institutions; it also provides some social benefits that enforcement discretion does not. Under expanded access the FDA would know who is doing FMT, for what purpose, and by what method. It could ensure that clinical protocols for performing FMT reflect up-to-date knowledge. The agency would also receive some adverse event reports related to the expanded access use.145 These features would make FMT safer for patients. Furthermore, expanded access for an intermediate-size patient population aims to minimize administrative burdens by permitting many patients to be treated under a single expanded-access IND or protocol.

This article's proposal places stool for FMT on a similar regulatory footing to stool-derived microbial products by removing much of the enforcement discretion now granted to stool. It also provides similar incentives for institutions to participate in or refer patients to studies of stool for FMT or studies of stool-derived microbial products. This more even regulatory approach should help secure investment in research and development for stool-derived microbial products.

Another salutary feature of this article's proposal is that it would retain much of the market for stool banks. Stool banks would not be regulatorily disadvantaged in comparison to in-house stool production, and could still distribute their products to some institutions under enforcement discretion (although this sector of the market would shrink considerably). Existing stool banks have played an important role in educating patients and advocating for them, and have played an important role in research on FMT.146 For these reasons, and because a product from stool banks likely is safer for some patients than in-house-produced stool, a regulatory policy should seek to retain stool banks. However, because the risks posed by centralized manufacturing and distribution of stool products are real, either the banks or the frequent recipients (heavy users) of their products should have INDs. The FDA should know who is producing stool for wide-spread distribution, how they are screening and tracking it, and to whom they are distributing it.

Governance involves more than regulation. Modern scholarship views governance as a polycentric activity in which the expertise to ensure adequate oversight is distributed among many actors.Reference Freeman, Dorf, Sabel, Lobel, Burris, Drahos, Shearing and Solomon147 For this and other reasons, scholars view agencies' engagement with stakeholders as a crucial tool of governance.Reference Lobel, Levi-Four and Trubek148 This article proposes that the FDA should undertake extensive and varied stakeholder engagement to generate ideas regarding RCT designs that are: (1) acceptable and attractive to patients and their clinicians; (2) capable of efficiently producing data the FDA will accept in support of a BLA for a stool-derived microbial product or for stool used in FMT; and (3) feasible and affordable.

To generate high-quality evidence, researchers and the FDA must find RCT designs that attract participants and clinicians. A new approach to regulatory enforcement will not promote evidence generation for FMT or stool-derived microbial products if patients turn to DIY FMT instead of clinical trials. Given patients' current enthusiasm for FMT, and practice guidelines recommending it as a treatment for rCDI, it seems likely that RCTs for both stool and stool-derived microbial products to treat rCDI will need to offer FMT to participants whose symptoms do not resolve during the trial. The obvious approach is to offer FMT as an open-label extension following the RCT portion of a trial.149 Open-label extensions could provide some information on the longer-term safety of FMT; however, their main purpose would be to encourage enrollment in RCTs. Also, such a trial design could slightly expand the market for products from stool banks. Open-label extension with FMT has been used in some current or recent RCTs.Reference Johnsen150

Stakeholder engagement might elicit other study design features that could attract participants to RCTs. For instance, a survey indicated that patients' most significant concern about FMT is disease transmission.151 RCTs that emphasize product screening and patient safety monitoring might, therefore, excel at accruing participants.

Stakeholder engagement should also help the FDA and research sponsors agree on trial designs that are efficient and likely to produce evidence that can support a BLA. They could address questions such as whether there are surrogate endpoints that could be measured in RCTs for FMT or stool-derived products. Engagements could help identify biomarkers or other tools that researchers in industry and academia should be trying to qualify.152 The use of biomarkers and valid surrogate endpoints could lead to more efficient production of high-quality evidence.

In addition to its merits, this article's proposal has some drawbacks. Most significantly, the FDA could generate powerful opposition by regulating in-house production of stool for FMT. Some organizations will argue that the FDA has no jurisdiction over in-house manufacturing and use. However, if stool produced by banks is regulated while stool produced in-house is not, then some (perhaps many) research-capable healthcare institutions will simply switch from using stool banks to producing stool in-house. These research-capable institutions would continue providing FMT as an unapproved treatment without generating evidence regarding its safety and efficacy.

Under the FDA's interpretation of its authorities, it has jurisdiction if a product meets the definition of a drug and is manufactured using materials obtained through interstate commerce. One federal appellate court and one federal district court have affirmed this legal interpretation with respect to production and use of stem cells.Reference Grady and Can153 Nonetheless, some health care organizations would view regulation of in-house stool production as agency overreach.

The FDA's approach to implementation would influence the degree of opposition to this article's proposed policy. For instance, before ceasing enforcement discretion the agency should provide enough time for investigators at research-capable healthcare organizations to find collaborators and initiate studies, begin referring patients to RCTs, and otherwise adjust to the coming policy change. Stakeholder engagements could help research-capable institutions better understand the proposed policy and help the FDA identify effective processes for implementation.

Conclusion

Good governance of products for microbiota transplants should encourage the production of high-quality evidence of their safety and efficacy, but the FDA's current and former regulatory policies have failed to achieve this goal for FMT. Despite its widespread use, patients and clinicians lack strong evidence for FMT's safety and efficacy for treating rCDI and other indications.

To the extent that particular combinations of microbes from donors cure patients' diseases, stool-derived microbial products containing those beneficial microbes likely will perform better as drug products than stool. Yet, the FDA's current and former regulatory policies may be undermining incentives for firms to invest in developing these products.

This article's governance proposal would decrease the FDA's use of enforcement discretion with regard to stool for FMT. The proposal is designed to change the way in which patients access FMT, routing patients to study participation or expanded access for intermediate-size patient populations. This proposal could produce more and higher-quality evidence for FMT, improve patient safety, and maintain incentives for private investment in the development of stool-derived microbial products. In theory, more evidence development should lead to quicker approval of some product or products that are safe and effective for treating rCDI, after which most patients could receive treatment without the need for expanded access.

Acknowledgements

The authors thank the Wisconsin Alumni Research Foundation and the Morgridge Institute for Research for salary support. We thank three peer reviewers for extremely helpful comments.

Footnotes

Dr. Ossorio reports personal fees from Genentech-Roche, outside the submitted work.

References

Proctor, L.M., “The Human Microbiome Project in 2011 and Beyond,” Cell Host & Microbe 10, no. 4 (2011): 287291.CrossRefGoogle Scholar
DeFilipp, Z., Hohmann, E., Jenq, R.R., and Chen, Y.B., “Fecal Microbiota Transplantation: Restoring the Injured Microbiome after Allogeneic Hematopoietic Cell Transplantation,” Biology of Blood and Marrow Transplantation 25, no. 1 (2019): e17e22.CrossRefGoogle Scholar
See Lee, W.J., Lattimer, L.D.N., Stephen, S., Borum, M.L., and Doman, D.B., “Fecal Microbiota Transplantation: A Review of Emerging Indications Beyond Relapsing Clostridum difficile Toxin Colitis,” Gastroenterology & Hepatology 11, no. 1 (2015): 2432; Mileto, S., Das, A., and Lyras, D., “Enterotoxic Clostridia: Clostridioides difficile Infections,” Micobiolical Spectrum 7, no. 3 (2019): GPP3-0015-2018.Google Scholar
de Vos, W.M. and de Vos, E.A., “Role of The Intestinal Microbiome in Health and Disease: From Correlation to Causation,” Nutrition Reviews 70, suppl. 1 (2012): S45S56.CrossRefGoogle Scholar
Bäckhed, F., Ding, H., Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A., Semenkovich, C.F., and Gordon, J.I., “The Gut Microbiota as An Environmental Factor That Regulates Fat Storage,” PNAS 101, no. 44 (2004): 1571815723; Turnbaugh, P.J., Ley, R.E., Mahowald, M.A., Magrini, V., Mardis, E.R., and Gordon, J.I., “An Obesity-Associated Gut Microbiome with Increased Capacity for Energy Harvest,” Nature 444, no. 7122 (2006): 10271031.CrossRefGoogle Scholar
See de Vos and de Vos, supra note 4; Quraishi, M.N., Widlak, M., Bhala, N., Moore, D., Price, M., Sharma, N., and Iqbal, T.H., “Systematic Review with Meta-Analysis: The Efficacy of Faecal Microbiota Transplantation for the Treatment of Recurrent and Refractory Clostridium difficile Infection,” Alimentary Pharmacology & Therapeutics 46, no. 5 (2017): 479493.CrossRefGoogle Scholar
Swaminath, A., “The Power of Poop: Patients Getting Ahead of Their Doctors Using Self-Administered Fecal Transplants,” The American Journal of Gastroenterology 109, no. 5 (2014): 777778; Jeffries, A., The World of Do-It-Yourself Fecal Transplants (Thanks, YouTube!) (December 8, 2014), Motherboard, available at <https://www.vice.com/en_us/article/3dk5w9/a-guide-to-do-it-yourself-fecal-transplants-thanks-youtube> (last visited September 11, 2019); D'Agostino, S., When an FDA Ruling Curbed Fecal Transplants, I Performed My Own (November 18, 2018), UNDark, available at <https://undark.org/2018/11/08/my-diy-fecal-transplant/> (last visited September 11, 2019).CrossRefGoogle Scholar
Ekekezie, C., Simmons, S., Perler, B., Burhke, T., Duff, C., Hott, B., Lillis, C.J., and Kelly, C.R., “Understanding the Scope of Do-It-Yourself Fecal Microbiota Transplantation (FMT),” American Journal of Gastroenterology 113, no. supp. S (2018): S102; Subbaraman, N., Shit Stirrers: Meet the People Doing Poop Transplants The Government Doesn't Want Them To, June 28, 2017, BuzzFeed.News, available at <https://www.buzzfeednews.com/article/nidhisubbaraman/inside-tampa-fecal-transplant-clinic> (last visited December 6, 2019); Khoruts, A., Hoffmann, D.E., and Palumbo, F.B., “The Impact of Regulatory Policies on the Future of FMT,” Journal of Law, Medicine & Ethics 47, no. 4 (2019): 482504CrossRefGoogle Scholar
Discussed infra, section I.Google Scholar
Lietzan, E., “Access before Evidence and the Price of the FDA's New Drug Authorities,” University of Richmond Law Review 53, no. 4 (2019): 12431309.Google Scholar
Molteni, M., “Patients Want Poop Transplants. Here's How to Make Them Safe,” Wired, December 14, 2017, available at <https://www.wired.com/story/patients-want-poop-transplants-heres-how-to-make-them-safe/> (last visited September 16, 2019).Google Scholar
Alang, N. and Kelly, C.R., “Weight Gain After Fecal Microbiota Transplantation,” Open Forum Infectious Diseases 2, no. 1 (2015): ofv004; Kelly, C.R., Ihunnah, C., Fischer, M., et al., “Fecal Microbiota Transplant for Treatment of Clostridium difficile Infection in Immunocompromised Patients,” American Journal of Gastroenterology 109, no. 7 (2014): 10651071; De Leon, L.M., Watson, J.B., and Kelly, C.R., “Transient Flare of Ulcerative Colitis after Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection,” Clinical Gastroenterology and Hepatology 11, no. 8 (2013): 10361038.CrossRefGoogle Scholar
U.S. Food and Drug Administration, Important Safety Alert Regarding Use of Fecal Microbiota for Transplantation and Risk of Serious Adverse Reactions Due to Transmission of Multi-Drug Resistant Organisms, FDA Website (June 13, 2019), FDA Website, available at <https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/important-safety-alert-regarding-use-fecal-microbiota-transplantationand-risk-serious-adverse> (last visited September 16, 2019).+(last+visited+September+16,+2019).>Google Scholar
Swaminath, A., “The Power of Poop: Patients Getting Ahead of Their Doctors Using Self-Administered Fecal Transplants,” The American Journal of Gastroenterology 109, no. 5 (2014): 777778. Discussed infra, note 67, 68, and accompanying text.CrossRefGoogle Scholar
Williams, J., The Updated List of Microbiome Companies and Investors, Global Engage, June 27, 2018, available at <http://www.global-engage.com/life-science/the-updated-list-ofmicrobiome-companies-and-investors/> (last visited September 16, 2019); Seres Therapeutics, Pipeline, Seres Website, available at <https://www.serestherapeutics.com/pipeline/products> (last visited September 16, 2019); Rebiotix, Pipeline, Rebiotix website, available at <https://www.rebiotix.com/rebiotix-product-pipeline/> (last visited September 16, 2019).Google Scholar
Discussed infra, section II.Google Scholar
See Eisenberg, R.S., “The Role of the FDA in Innovation Policy,” Michigan Telecommunications and Technology Law Review 13, no. 2 (2007): 345388; Kapczynski, A., “Dangerous Times: The FDA's Role in Information Production, Past and Future,” Minnesota Law Review 102, no. 6 (2018): 23572382; Lietzan, supra note 10.Google Scholar
Lietzan, supra note 10.Google Scholar
U.S. Food and Drug Administration, Guidance for Industry: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies (July 2013), available at <https://www.fda.gov/downloads/BiologicsBloodVaccines/Guidance-ComplianceRegulatoryInformation/Guidances/Vaccines/UCM361393.pdf> (last visited September 16, 2019).+(last+visited+September+16,+2019).>Google Scholar
Discussed infra, section III.Google Scholar
Many commentators believe that the next big opportunity for microbiome-based product development will be in oncology because certain microbes have been associated with resistance to antitumor drugs while other microbes appear necessary for the effective functioning of some antitumor drugs. McQuade, J.L., Daniel, C.R., Helmink, B.A., and Wargo, J.A., “Modulating the Microbiome to Improve Therapeutic Response to Cancer,” The Lancet: Oncology 20, no. 2 (2019): 7791; Routy, B., Gopalakrishnan, V., Daillère, R., et al., “The Gut Microbiota Influences Anticancer Immunosurveillance and General Health,” Nature Reviews Clinical Oncology 15, no. 6 (2018): 382396; Zitvogel, L., Ayyoub, M., Routy, B., and Kroemer, G., “Microbiome and Anticancer Immunosurveil-lance,” Cell 165, no. 2 (2016): 267287.CrossRefGoogle Scholar
See Eisenberg, supra note 17; Lietzan, supra note 10.Google Scholar
Lietzan, supra note 10, at 1300–1309.Google Scholar
See Panchal, P., Budree, S., Scheeler, A., et al., “Scaling Safe Access to Fecal Microbiota Transplantation: Past, Present, and Future,” Current Gastroenterology Reports 20, no. 4 (2018): 14.CrossRefGoogle Scholar
Friedman, L.M., Furberg, C.D., DeMets, D.L., et al., Fundamentals of Clinical Trials (Switzerland: Springer International Publishing, 2015): chapter 11.CrossRefGoogle Scholar
Fisher, R., “Cigarettes, Cancer, and Statistics,” The Centennial Review of Arts & Science 2 (1958): 151166; Bothwell, L.E., Greene, J.A., Podolsky, S.H., and Jones, D.S., “Assessing the Gold Standard — Lessons from the History of RCTs,” The New England Journal of Medicine 374, no. 22 (2016): 21752181. Prior to conducting an RCT, a sponsor typically conducts smaller, and sometimes unblinded or uncontrolled trials to elicit information about the investigational drug's pharmaco-dynamics, pharmacokinetics, dosing, safety, and tolerability. These preliminary trials can also provide preliminary evidence of efficacy. See Friedman, et al., supra note 25, at 4–5.Google Scholar
Kendall, J.M., “Designing a Research Project: Randomized Controlled Trials and their Principles,” Emergency Medical Journal 20, no. 2 (2003): 164168; Friedman, L.M., Furberg, C.D., DeMets, D.L., et al., Fundamentals of Clinical Trials (Switzerland: Springer International Publishing, 2015).Google Scholar
Randomization consists of “generating an unpredictable random sequence” and then implementing it in a manner that conceals which group research participants have been assigned to. Dettori, J., “The Random Allocation Process: Two Things You Need to Know,” Evidence-based Spine-Care Journal 1, no. 3 (2010). To conduct a properly randomized trial, people in both (or all) arms should be treated identically except for the difference in administering the test or control interventions. Prior to randomization, researchers should measure known prognostic and risk factors relating to the disease under study and the outcome of interest (potential confounding factors). At the analysis stage, investigators should demonstrate that potential confounding factors were evenly distributed among people in each arm of the trial or that the investigators made appropriate statistical adjustments for confounding. Bothwell, L.E., Greene, J.A., Podolsky, S.H., and Jones, D.S., “Assessing the Gold Standard — Lessons from the History of RCTs,” The New England Journal of Medicine 374, no. 22 (2016): 21752181; Kendall, J.M., “Designing a Research Project: Randomized Controlled Trials and their Principles,” Emergency Medical Journal 20, no. 2 (2003): 164168.CrossRefGoogle Scholar
Blinding simply means that a person does not know whether any participant is in the control or test arm of a study. When a study is described as single-blinded, it generally means the participants do not know whether they are in an experimental or control arm of the study, but the investigators do know. However, most commentators view studies in which both the investigators and the participants are blinded as being more reliable. Numerous empirical studies have shown a substantial degree of bias associated with failure to blind the researchers who conduct a study or analyze its outcomes. Podolsky, S.H., Jones, D.S., and Kaptchuk, T.J., “From Trials to Trials: Blinding, Medicine, and Honest Adjudication,” in Kesselheim, A.S. and Robertson, C.T., eds., Blinding as a Solution to Bias: Strengthening Biomedical Science, Forensic Science, and Law (Academic Press, 2016): at 46; Hróbjartsson, A., “Blinding in Biomedical Research: An Essential Method to Reduce Risk of Bias,” in Kesselheim, A.S. and Robertson, C.T., eds., Blinding as a Solution to Bias: Strengthening Biomedical Science, Forensic Science, and Law (Academic Press, 2016): at 60, 63–66; Begley, C.G. and Ioannidis, J.P., “Reproducibility in Science: Improving the Standard for Basic and Preclinical Research,” Circulation Research 116, no. 1 (2015): 116126.Google Scholar
U.S. Food and Drug Administration, Guidance for Industry: E10 Choice of Control Group and Related Issues in Clinical Trials, FDA website (May 2001), available at <https://www.fda.gov/media/71349/download> (last visited September 16, 2019). Here, bias means that estimates of a treatment's effect systematically deviate from its true value. Id. at 3.+(last+visited+September+16,+2019).+Here,+bias+means+that+estimates+of+a+treatment's+effect+systematically+deviate+from+its+true+value.+Id.+at+3.>Google Scholar
Friedman, et al., supra note 25, at 31–33.Google Scholar
International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use, Choice of Control Group and Related Issues in Clinical Trials, July 2000 (noting that the choice of control group “affects the inferences that can be drawn from the trial, the ethical acceptability of the trial, the degree to which bias in conducting and analyzing the study can be minimized, the types of subjects that can be recruited and the pace of recruitment, the kinds of endpoints that can be studied…” at 1; Bothwell, L.E., Greene, J.A., Podolsky, S.H., and Jones, D.S., “Assessing the Gold Standard — Lessons from the History of RCTs,” The New England Journal of Medicine 374, no. 22 (2016): 21752181; Friedman, et al., supra note 25, at 31–32 (noting that “a blinded placebo control provides the most complete information about the risks and benefits of a new therapy as an inert placebo is the best approximation of a neutral control.”).Google Scholar
See Sherman, R.E., Anderson, S.A., Dal Pan, G.J., et al., “Real-World Evidence — What Is It and What Can It Tell Us?” The New England Journal of Medicine 375, no. 23 (2016): 22932297; see also, Ejima, K., Li, P., Smith, D.L. Jr., et al., “Observational Research Rigor Alone Does Not Justify Causal Inference,” European Journal of Clinical Investigation 46, no. 12 (2016): 985993.CrossRefGoogle Scholar
Hormone replacement therapy (HRT) for post-menopausal women provides an excellent example of how observational research can mislead. In the 1980s and early 1990s, observational studies suggested that post-menopausal women benefited from HRT and that there were few risks associated with this intervention. See Henderson, B.E., Paganini-Hill, A., and Ross, R.K., “Decreased Morality in Users of Estrogen Replacement Therapy,” Archives of Internal Medicine 151, no. 1 (1991): 7578; Stampfer, M.J. and Colditz, G.A., “Estrogen Replacement Therapy and Coronary Heart Disease: A Quantitative Assessment of the Epidemiologic Evidence,” Preventive Medicine 20, no. 1 (1991): 4763; Grady, D., Rubin, S.M., Petitti, D.B., et al., “Hormone Therapy to Prevent Disease and Prolong Life in Postmenopausal Women,” Annals of Internal Medicine 117, no. 12 (1992): 10161037. There was a biologically plausible explanation for why women should benefit from HRT. However, in the 1990s a prospective, randomized, double-blinded placebo-controlled, clinical trial involving over 16,000 women showed that the benefits of HRT were smaller and fewer than people believed, and the risks were greater. The RCT was stopped early because the treatment group's incidence of invasive breast cancer exceeded the rate set by the data and safety monitoring board's prearranged stopping rule, and the statistic summarizing risks as compared to benefits indicated that the risks of HRT exceeded its benefits in the study population. Hulley, S., Grady, D., Bush, T., et al., for the Heart and Estrogen/Progestin Replacement Study (HERS) Research Group, “Randomized Trial of Estrogen Plus Progestin for Secondary Prevention of Coronary Heart Disease in Postmenopausal Women,” JAMA 280, no. 7 (1998): 605613; Writing Group for the Women's Health Initiative Investigators, “Risks and Benefits of Estrogen Plus Progestin in Healthy Postmenopausal Women: Principal Results from the Women's Health Initiative Randomized Controlled Trial,” JAMA 288, no. 3 (2002): 321333; Writing Group for the Women's Health Initiative Investigators, “Risks and Benefits of Estrogen Plus Progestin in Healthy Post-menopausal Women: Principal Results from the Women's Health Initiative Randomized Controlled Trial,” JAMA 288, no. 3 (2002): 321333.CrossRefGoogle Scholar
See, e.g., U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Manual of Policies and Procedures 4000.4 Rev. 1, Good Review Practices: Clinical Pharmacology Review of New Molecular Entity (NME) New Drug Applications (NDAs) and Original Biologics License Applications (BLAs) (Sept. 23, 2016), available at <https://www.fda.gov/media/71709/download> (stating that “Reviews may include independent OCP data analysis.”) (last visited September 16, 2019); U.S. Food and Drug Administration, Drug Development and Review Definitions, FDA website, available at <https://www.fda.gov/drugs/investigational-new-drug-ind-application/drug-development-and-review-definitions> (“The review is likely to involve a reanalysis or an extension of the analyses performed by the sponsor and presented in the NDA.”) (last visited September 16, 2019).+(stating+that+“Reviews+may+include+independent+OCP+data+analysis.”)+(last+visited+September+16,+2019);+U.S.+Food+and+Drug+Administration,+Drug+Development+and+Review+Definitions,+FDA+website,+available+at++(“The+review+is+likely+to+involve+a+reanalysis+or+an+extension+of+the+analyses+performed+by+the+sponsor+and+presented+in+the+NDA.”)+(last+visited+September+16,+2019).>Google Scholar
Lietzan, supra note 10, at 1301 (noting that the FDA receives an average of 70 to 80 new drug applications per year, and data in support of each application can run to a half million or more pages, which the FDA reviews at an average cost of over $1.2 million per application.).Google Scholar
Kapczynski, supra note 17. Discussed in more detail in Section III, infra.Google Scholar
Kelly, C.R., Kim, A.M., Laine, L., and Wu, G.D., “The AGA's Fecal Microbiota Transplantation National Registry: An Important Step Toward Understanding Risks and Benefits of Microbiota Therapeutics,” Gastroenterology 152, no. 4 (2017): 681684.CrossRefGoogle Scholar
Allegretti, J.R., Kassam, Z., Osman, M., et al., “The 5D Framework: A Clinical Primer for Fecal Microbiota Transplantation to Treat Clostridium difficile Infection,” Gastrointestinal Endoscopy 87, no. 1 (2018): 1829; Kelly, C.R., Khoruts, A., Staley, C., et al., “Effect of Fecal Microbiota Transplantation on Recurrence in Multiply Recurrent Clostridium difficile Infection,” Annals of Internal Medicine 165, no. 9 (2016): 609616.Google Scholar
U.S. Centers for Disease Control and Prevention, Nearly Half a Million Americans Suffered from Clostridium difficile Infections in a Single Year (February 25, 2015), available at <https://www.cdc.gov/media/releases/2015/p0225-clostridium-difficile.html> (last visited September 16, 2019).+(last+visited+September+16,+2019).>Google Scholar
Lessa, F.C., Mu, Y., Bamberg, W.M., et al., “Burden of Clostridium difficile Infection in the United States,” The New England Journal of Medicine 372, no. 24 (2015): 23692370; see GlobalData, Global Clostridium difficile Infections Market To Approach $1.7 Billion By 2026 (July 24, 2017), available at <https://www.globaldata.com/global-clostridium-difficile-infections-market-approach-1-7-billion-2026/> (last visited September 16, 2019); Edelstein, C., Daw, J.R., and Kassam, Z., “Seeking Safe Stool: Canada Needs a Universal Donor Model,” CMAJ 188, no. 17/18 (2016): 1718.CrossRefGoogle Scholar
Scheeler, A., “Where Stool is a Drug: International Approaches to Regulating the Use of Fecal Microbiota for Transplantation,” Journal of Law, Medicine & Ethics 47, no. 4 (2019): 524540, Table 1.CrossRefGoogle Scholar
Quraishi, M.N., Widlak, M., Bhala, N., et al., “Systematic Review with Meta-Analysis: The Efficacy of Faecal Microbiota Transplantation for the Treatment of Recurrent and Refractory Clostridium difficile Infection,” Alimentary Pharmacology & Therapeutics 46, no. 5 (2017): 479493; Gough, E., Shaikh, H., and Manges, A.R., “Systematic Review of Intestinal Microbiota Transplantation (Fecal Bacteriotherapy) for Recurrent Clostridium difficile Infection,” Clinical Infectious Diseases 53, no. 10 (2011): 9941002; Drekonja, D., Reich, J., Gezahegn, S., et al., “Fecal Microbiota Transplantation for Clostridium difficile Infection: A Systematic Review,” Annals of Internal Medicine 162, no. 9 (2015): 630638; Kassam, Z., Lee, C.H., and Hunt, R.H., “Review of the Emerging Treatment of Clostridium difficile Infection with Fecal Microbiota Transplantation and Insights into Future Challenges,” Clinics in Laboratory Medicine 34, no. 4 (2014): 787798.CrossRefGoogle Scholar
Kelly, C.R., Kahn, S., Kashyap, P., et al., “Update on Fecal Microbiota Transplantation 2015: Indications, Methodologies, Mechanisms and Outlook,” Gastroenterology 149, no. 1 (2015): 223237.CrossRefGoogle Scholar
van Nood, E., Vrieze, A., Nieuwdorp, M., et al., “Duedenal Infusion of Donor Feces for Recurrent Clostridium difficile,” The New England Journal of Medicine 368, no. 5 (2013): 407415.Google Scholar
Kelly et al., supra note 40.Google Scholar
See, e.g., Seekatz, A.M., Rao, K., Santhosh, K., and Young, V.B., “Dynamics of the Fecal Microbiome in Patients with Recurrent and Nonrecurrent Clostridium difficile Infection,” Genome Medicine 8, no. 1 (2016): at 47 (“The success of microbial-based therapeutics, such as fecal microbiota transplantation, for the treatment of recurrent CDI underscores the importance of restoring the microbiota.”).Google Scholar
National Institutes of Health, Clinical Trial Testing Fecal Microbiota Transplant for Recurrent Diarrheal Disease Begins, NIH website (January 14, 2019), available at <https://www.niaid.nih.gov/news-events/clinical-trial-testing-fecalmicrobiota-transplant-recurrent-diarrheal-disease-begins> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
Clinicaltrials.gov, Safety and Efficacy of FMT in Individuals with One or More Recurrences of Clostridium difficile Associated Disease (CDAD), Identifier No. NCT3548051, posted June 6, 2018, updated September 2, 2019, available at <https://clinicaltrials.gov/ct2/show/NCT03548051> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
See, e.g., Infectious Disease Society of America, Catherine Duff: A Woman Undergoes Fecal Microbiota Transplant (FMT) to Cure Her Recurring Clostridium difficile Infections, Saving Her Life, available at <https://www.idsociety.org/public-health/patient-stories/catherine-duff/> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
McDonald, L.C., Gerding, D.N., Johnson, S., et al., “Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: 2017 Update by the Infectious Disease Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA),” Clinical Infectious Diseases 66, no. 7 (2018): e1e48.CrossRefGoogle Scholar
Aetna, Fecal Bacteriotherapy (November 29, 2018), Clinical Policy Bulletin Number 0844, available at <http://www.aetna.com/cpb/medical/data/800_899/0844.html> (last visited September 17, 2019); HealthNet, Clinical Policy: Fecal Bacteriotherapy, Reference No. HNCA.CP.MP.519 (effective November, 2016), HealthNet Website, available at <https://www.healthnet.com/static/general/unprotected/pdfs/.../FecalBacteriotherapy.pdf>(last visited September 17, 2019); BlueCross BlueShield of North Carolina, Corporate Medical Policy: Fecal Microbiota Transplantation, BlueCrossNC Website, available at <https://www.bluecrossnc.com/sites/default/files/document/attachment/services/public/pdfs/medicalpolicy/fecal_microbiota_transplantation.pdf> (last visited September 17, 2019). Note that payers' definitions of rCDI may differ with respect to how many recurrences are necessary, or what kinds of previous treatments need to have failed prior to undertaking FMT. In contrast, OpenBiome has stated that many private insurers do not pay for FMT for rCDI, and the organization has initiated fundraising activities to help patients pay for stool and FMT. See OpenBiome, OpenBiome is Seeking Input on a Draft Proposal to FDA about Regulation of FMT (November 7, 2018), OpenBiome website, available at <https://www.openbiome.org/comment-to-fda> (last visited September 17, 2019).+(last+visited+September+17,+2019);+HealthNet,+Clinical+Policy:+Fecal+Bacteriotherapy,+Reference+No.+HNCA.CP.MP.519+(effective+November,+2016),+HealthNet+Website,+available+at+(last+visited+September+17,+2019);+BlueCross+BlueShield+of+North+Carolina,+Corporate+Medical+Policy:+Fecal+Microbiota+Transplantation,+BlueCrossNC+Website,+available+at++(last+visited+September+17,+2019).+Note+that+payers'+definitions+of+rCDI+may+differ+with+respect+to+how+many+recurrences+are+necessary,+or+what+kinds+of+previous+treatments+need+to+have+failed+prior+to+undertaking+FMT.+In+contrast,+OpenBiome+has+stated+that+many+private+insurers+do+not+pay+for+FMT+for+rCDI,+and+the+organization+has+initiated+fundraising+activities+to+help+patients+pay+for+stool+and+FMT.+See+OpenBiome,+OpenBiome+is+Seeking+Input+on+a+Draft+Proposal+to+FDA+about+Regulation+of+FMT+(November+7,+2018),+OpenBiome+website,+available+at++(last+visited+September+17,+2019).>Google Scholar
Centers for Medicare and Medicaid Services, CMS Manual System Pub 100-04: Medicare Claims Processing, CMS Website (December 22, 2017), available at <https://www.cms.gov/Regulations-and-Guidance/Guidance/Transmittals/2017Downloads/R3940CP.pdf> (last visited September 17, 2019). Medicare is a particularly important source of funding because most patients with CDI are 65 years of age or older. Lessa, F.C., Mu, Y., Bamberg, W.M., et al., “Burden of Clostridium difficile Infection in the United States,” The New England Journal of Medicine 372, no. 24 (2015): at 828.Google Scholar
See Vrieze, A., de Groot, P.F., Kootte, R.S., et al., “Fecal Transplant: A Safe and Sustainable Clinical Therapy for Restoring Intestinal Microbial Balance in Human Disease?” Best Practice & Research Clinical Gastroenterology 27, no. 1 (2013): 127137; Kelly et al., supra note 45.Google Scholar
310 Studies Found for “Fecal Transplant,” available at <https://clinicaltrials.gov/ct2/results?cond=&term=fecal+transplant&cntry=&state=&city=&dist=> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
See U.S. Food and Drug Administration, Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease: Guidance for Industry (October 2018), available at <https://www.fda.gov/media/117173/download> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
See Sherman, R.E., Anderson, S.A., Dal Pan, G.J., et al., “Real-World Evidence — What Is It and What Can It Tell Us?” The New England Journal of Medicine 375, no. 23 (2016): 22932297; Network for Excellence in Health Innovation, Real World Evidence: A New Era for Health Care Innovation (September 2015), available at <https://www.nehi.net/writable/publication_files/file/rwe_issue_brief_final.pdf>; National Academies of Science, Engineering, and Medicine, Examining the Impact of Real-World Evidence on Medical Product Development: Proceedings of a Workshop Series, 2019.Google Scholar
National Academies of Science, Engineering, and Medicine, Examining the Impact of Real-World Evidence on Medical Product Development: Proceedings of a Workshop Series, 2019.Google Scholar
Edelstein, C.A., Kassam, Z., Daw, J., et al., “The Regulation of Fecal Microbiota for Transplantation: An International Perspective for Policy and Public Health,” Clinical Research and Regulatory Affairs 32, no. 3 (2015): 101109.CrossRefGoogle Scholar
Kelly et al., supra note 39.Google Scholar
Sherman, R.E., Anderson, S.A., Dal Pan, G.J., et al., “Real-World Evidence — What Is It and What Can It Tell Us?” The New England Journal of Medicine 375, no. 23 (2016): 22932297; Network for Excellence in Health Innovation, Real World Evidence: A New Era for Health Care Innovation (September 2015), available at <https://www.nehi.net/writable/publication_files/file/rwe_issue_brief_final.pdf> (last visited September 17, 2019); Pearl, J., Causality: Models, Reasoning, and Inference (New York: Cambridge University Press, 2009): at 350; McMurray, J., “Only Trials Tell the Truth About Treatment Effects,” Journal of the American College of Cardiology 71, no. 23 (2018): 26402642; Freemantle, N., Marston, L., Walters, K., et al., “Making Inferences on Treatment Effects from Real World Data: Propensity Scores, Confounding by Indication, and Other Perils for the Unwary in Observational Research,” BMJ 347 (2013): f6409; see also National Academies of Science, Engineering, and Medicine, Examining the Impact of Real-World Evidence on Medical Product Development: Proceedings of a Workshop Series, 2019, at 1, 11, 23.CrossRefGoogle Scholar
Berger, M.L., Sox, H., Wilke, R.J., et al., “Good Practices for Real-World Data Studies of Treatment and/or Comparative Effectiveness: Recommendations from the Joint ISPORISPE Special Task Force on Real-World Evidence in Health Care Decision Making,” Value in Health 20, no. 8 (2017): 10031008.Google Scholar
See Kuang, Z., Bao, Y., Thomson, J., et al., “A Machine-Learning-Based Drug Repurposing Approach Using Baseline Regularization,” in Vanhaelen, Q., ed., Computational Methods for Drug Repurposing (New York: Humana Press, 2019): 255267; OHDSI, Frequently Asked Questions, available at <https://ohdsi.org/who-we-are/frequently-asked-questions/> (last visited September 17, 2019).CrossRefGoogle ScholarPubMed
See National Academies of Science, Engineering, and Medicine, Examining the Impact of Real-World Evidence on Medical Product Development: Proceedings of a Workshop Series, 2019, at 1; Gottlieb, S., Statement from FDA Commissioner Scott Gottlieb, M.D., on FDA's New Strategic Framework to Advance Use of Real-world Evidence to Support Development of Drugs and Biologics (December 6, 2018), available at <https://www.fda.gov/news-events/press-announcements/statement-fda-commissioner-scott-gottlieb-md-fdas-new-strategic-framework-advance-use-real-world> (September 17, 2019).Google Scholar
In a previous article, the authors discussed the safety of FMT. Ossorio, P.N. and Zhou, Y., “Regulating Stool for Microbiota Transplantation,” Gut Microbes 10, no. 2 (2019): 105108.CrossRefGoogle Scholar
Kelly et al., supra note 45; Baxter, M. and Colville, A., “Adverse Events in Fecal Microbiota Transplant: A Review of the Literature,” Journal of Hospital Infection 92, no. 2 (2016): 117127.Google Scholar
Kelly et al., supra note 45; Kelly et al., supra note 39.Google Scholar
See Baxter and Colville, supra note 68; Qazi, T., et al., “The Risks of Inflammatory Bowel Disease Flares after Fecal Microbiota Transplantation: Systematic Review and Meta-analysis,” Gut Microbes 8, no. 6 (2017): 574588.CrossRefGoogle Scholar
Paramsothy, S., Borody, T.J., Lin, E., et al., “Donor Recruitment for Fecal Microbiota Transplantation,” Inflammatory Bowel Disease 21, no. 7 (2015): 16001606; Park, L., Mone, A., Price, J.C., et al., “Perceptions of Fecal Microbiota Transplantation for Clostridium difficile Infection: Factors that Predict Acceptance,” Annals of Gastroenterology 30, no. 1 (2017): 8388.CrossRefGoogle Scholar
Dubois, N., Ling, K., Osman, M., et al., “Prospective Assessment of Donor Eligibility for Fecal Microbiota Transplantation at a Public Stool Bank: Results from the Evaluation of 1387 Candidate Donors,” Open Forum Infectious Diseases 2, no. supp 1 (2015): at 962; Paramsothy, et al., supra note 71; Edelstein, et al., supra note 42.CrossRefGoogle Scholar
Some organizations or collections have highly regarded professionals have proposed standards. Reviewed in Wood-worth, M.H., Neish, E.M., Miller, N.S., et al., “Laboratory Testing of Donors and Stool Samples for Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection,” Journal of Clinical Microbiology 55, no. 4 (2017): 10021010; see also, Terveer, E.M., van Beurden, Y.H., Goorhuis, A., et al., “How to: Establish and Run a Stool Bank,” Clinical Microbiology and Infection 23, no. 12 (2017): 924930.CrossRefGoogle Scholar
FDA scientists have recently announced requirements for testing stool for multi-drug resistant organisms (MDROs). U.S. Food and Drug Administration, supra note 13.Google Scholar
Dubois, N., Ling, K., Osman, M., et al., “Prospective Assessment of Donor Eligibility for Fecal Microbiota Transplantation at a Public Stool Bank: Results from the Evaluation of 1387 Candidate Donors,” Open Forum Infectious Diseases 2, no. supp. 1 (2015): at 962; Paramsothy et al.,, supra note 71; Edelstein et al., supra note 42.CrossRefGoogle Scholar
See Ossorio and Zhou, supra note 67.Google Scholar
U.S. Food and Drug Administration, supra note 13.Google Scholar
21 C.F.R. § 312.Google Scholar
U.S. Food and Drug Administration, supra note 13.Google Scholar
Hoffmann, D.E., Palumbo, F.B., Ravel, J., et al., “A Proposed Definition of Microbiota Transplantation for Regulatory Purposes,” Gut Microbes 8, no. 3 (2017): 208213.CrossRefGoogle Scholar
Panchal, et al., supra note 24.Google Scholar
Id. Note that the first stool bank began shipping stool for FMT in 2012, four years before the first double-blinded, placebo-controlled RCT for rCDI was published. Lietzan, supra note 10, at 1296.Google Scholar
This article follows the definition in the FDA's 2016 draft guidance. U.S. Food and Drug Administration, Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies, Draft Guidance for Industry (March 2016), available at <https://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/Vaccines/UCM488223.pdf> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
The Human Microbiome Consortium, “Structure, Function, and Diversity of the Healthy Human Microbiome,” Nature 486, no. 7402 (2012): 207214.CrossRefGoogle Scholar
Lloréns-Rico, V. and Raes, J., “Tracking Humans and Microbes,” Nature 569, no. 7758 (2019): 632633.CrossRefGoogle Scholar
U.S. Food and Drug Administration, FDA 101: Regulating Biological Products (July 25, 2008), available at <https://www.fda.gov/consumers/consumer-updates/fda-101-regulating-biological-products> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
U.S. Food and Drug Administration, Frequently Asked Questions About Therapeutic Biologic Products, FDA Website, available at <https://www.fda.gov/drugs/therapeutic-biologics-applications-bla/frequently-asked-questions-about-therapeutic-biological-products> (last visited September 17, 2019); Lietzan, supra note 10, at 1265–1266.+(last+visited+September+17,+2019);+Lietzan,+supra+note+10,+at+1265–1266.>Google Scholar
Slater, J., U.S. Food and Drug Administration, Remarks at Fecal Microbiota Transplantation: Scientific and Regulatory Issues, transcript (May 3, 2013), available at <http://way-back.archive-it.org/7993/20170113133557/http://www.fda.gov/downloads/BiologicsBloodVaccines/NewsEvents/Work-shopsMeetingsConferences/UCM352902.pdf> (last visited September 17, 2019).Google Scholar
Hoffmann, et al., supra note 80.Google Scholar
The Integrative HMP (iHMP) Research Network Consortium, “The Integrative Human Microbiome Project,” Nature 569, no. 7758 (2019): 641648.CrossRefGoogle Scholar
See Sokol, H., Leducq, V., Aschard, H., et al., “Fungal Micro-biota Dysbiosis in IBD,” Gut 66, no. 6 (2017): 10391048; Chehoud, C., Dryga, A., Hwang, Y., et al., “Transfer of Viral Communities between Human Individuals during Fecal Micro-biota Transplantation,” mBIO 7, no. 2 (2016): e00322v–16; Lloyd-Price, J., Arze, C., Ananthakrishnan, A.N., et al., “Multi-omics of the Gut Microbial Ecosystem in Inflammatory Bowel Diseases,” Nature 569, no. 7758 (2019): 655662.CrossRefGoogle Scholar
See, Association for Molecular Pathology v. Myriad, 133 S.Ct. 2107 (2013).Google Scholar
See, e.g., Stevens, C.J., “Fecal Microbiome Transplant Material Preparation Method and Apparatus,” U.S. Patent No. 9,192,361, November 24, 2015; Sandowsky, M.J., Khoruts, A., Weingarden, A.R., and Hamilton, M.J., “Compositions and Methods for Transplantation of Colon Microbiota,” U.S. Patent No. 9,968,638, May 15, 2018.Google Scholar
NIH Human Microbiome Portfolio Analysis Team, “A Review of 10 Years of Human Microbiome Research Activities at the US National Institutes of Health, Fiscal Years 2007-2016,” Microbiome 7, no. 1 (2019): at 31; Gertner, J., What's Lurking in Your Microbiome? Possibly a Cure for Disease, Fast Company, January 7, 2015, available at <http://www.fastcompany.com/3039891/gut-check> (last visited September 17, 2019; a version of this article appeared in print in the February 2015 print edition of Fast Company magazine).Google Scholar
Hoffmann, et al., supra note 80.Google Scholar
Lietzan, supra note 10. See also Khoruts, et al., supra note 8.Google Scholar
Ossorio and Zhou, supra note 67. But see Khoruts, et al., supra note 8 (taking issue with assumption that defined consortia products will necessarily be superior to “complete community” products).Google Scholar
Khoruts et al., supra note 8.Google Scholar
DiMasi, J.A. and Grabowski, H.G., “R&D Costs and Returns to New Drug Development: A Review of the Evidence,” in Danzon, P.M. and Nicholson, S., eds., The Oxford Handbook of the Economics of the Biopharmaceutical Industry (Oxford University Press, 2012): at 21.Google Scholar
Lietzan, supra note 10, at 1304–1305.Google Scholar
See, e.g., U.S. Food and Drug Administration, Guidance for Industry: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies (July 2013), available at <https://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/Vaccines/UCM361393.pdf> (last visited September 17, 2019); U.S. Food and Drug Administration, Early Clinical Trials with Live Biotherapeutic Products: Chemistry, Manufacturing, and Control Information (updated June 2016), available at <https://www.fda.gov/media/82945/download> (last visited September 17, 2019).+(last+visited+September+17,+2019);+U.S.+Food+and+Drug+Administration,+Early+Clinical+Trials+with+Live+Biotherapeutic+Products:+Chemistry,+Manufacturing,+and+Control+Information+(updated+June+2016),+available+at++(last+visited+September+17,+2019).>Google Scholar
21 U.S.C. § 355(d).Google Scholar
21 U.S.C. §§ 356(a), 356(b), 356(c); U.S. Food and Drug Administration, Guidance for Industry: Expedited Programs for Serious Conditions – Drugs and Biologics (May 2014), available at <https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm358301.pdf> (last visited September 18, 2019); Lietzan, supra note 10, at 1266–1267.+(last+visited+September+18,+2019);+Lietzan,+supra+note+10,+at+1266–1267.>Google Scholar
For a description of the new drug approval process and FDA's post-market authorities see, Lietzan, supra note 35, ch. 4.; Lietzan, supra note 10, at 1280–1293.Google Scholar
Kapczynski, supra note 17; Lietzan, supra note 10, at 1268–1271.Google Scholar
Kapczynski, supra note 17, at 2364–2365 (Discussing the case of Vioxx, a non-steroidal anti-inflammatory drug. Merck had early evidence of serious cardiovascular side-effects but it did not include cardiovascular risk assessments in any of the clinical trial data it submitted to the FDA as part of its new drug application.).Google Scholar
21 C.F.R. § 312.Google Scholar
Lietzan, supra note 10, at 1272–1273.Google Scholar
21 C.F.R. § 312.80. Expanded access is a program that provides an investigational medical product to a patient who has an immediately life-threatening condition or serious disease or condition, when no comparable or satisfactory alternative therapy options are available. This access is granted outside of a clinical trial. U.S. Food and Drug Administration, Expanded Access, FDA Website, available at <https://www.fda.gov/NewsEvents/PublicHealthFocus/ExpandedAccessCompassionateUse/ucm20080392.htm> (last visited September 18, 2019).+(last+visited+September+18,+2019).>Google Scholar
Wendler, T., Mongiello, F., McLinn, J., and Bellina, M., Right to Try Act, P.L. 115176 (2018); U.S. Food and Drug Administration, Right to Try (May 28, 2019), available at <https://www.fda.gov/patients/learn-about-expanded-access-and-other-treatment-options/right-try> (last visited September 18, 2019).Google Scholar
Fischer, M., Kassam, Z., Kelly, C.R., and Sobcinski, M.K., Fecal Microbiota Transplantation: Moving Mainstream, Regulatory Hurdles and Potential Future Indication (June 2015), Healio Gastroenterology, available at <https://www.healio.com/gastroenterology/infection/news/print/healio-gastroenterology/%7Bc0da87d3-b483-40b7-846b-0777c2a76ec3%7D/fecal-microbiota-transplantation-moving-mainstream-regulatory-hurdles-and-potential-future-indications?page=3> (last visited September 18, 2019).Google Scholar
U.S. Food and Drug Administration, Guidance for Industry: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies (July 2013), available at <https://www.fda.gov/downloads/BiologicsBloodVaccines/Guidance-ComplianceRegulatoryInformation/Guidances/Vaccines/UCM361393.pdf> (last visited September 18, 2019). For an explanation of executive branch enforcement discretion see, Price, Z.S., “Enforcement Discretion and the Political Maelstrom,” Administrative and Regulatory Law News 42, no. 3 (2017): 1720; Price, Z.S., “Enforcement Discretion and Executive Duty,” Vanderbilt Law Review 67, no. 3 (2014): 671769.Google Scholar
OpenBiome asserts that it shipped 43,000 stool preparations as of January 2019. OpenBiome, Our Impact (January, 2019), OpenBiome Website, available at <https://www.openbiome.org/impact> (last visited September 18, 2019). Because some patients may have received more than one FMT using Open-Biome stool, and other patients have been treated using stool produced in-house by healthcare organizations, it is unclear how many patients have undergone FMT.+(last+visited+September+18,+2019).+Because+some+patients+may+have+received+more+than+one+FMT+using+Open-Biome+stool,+and+other+patients+have+been+treated+using+stool+produced+in-house+by+healthcare+organizations,+it+is+unclear+how+many+patients+have+undergone+FMT.>Google Scholar
Rebiotix, Comment on the Food and Drug Administration (FDA) Notice: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies: Draft Guidance for Industry (June 20, 2016), Regulations.gov Website, available at <https://www.regulations.gov/document?D=FDA-2013-D-0811-0116> (last visited September 18, 2019).+(last+visited+September+18,+2019).>Google Scholar
Hoffmann, D., Palumbo, F., Ravel, J., et al., “Improving Regulation of Microbiota Transplants,” Science 358, no. 6369 (2017): 13901391; Smith, M.B., Kelly, C., and Alm, E.J., “How to Regulate Faecal Transplants,” Nature 506, no. 7488 (2014): 290291; Sachs, R.E. and Edelstein, C.A., “Ensuring the Safe and Effective FDA Regulation of Fecal Microbiota Transplantation,” Journal of Law and the Biosciences 396, no. 2 (2015): 396415; Riley, M.F. and Olle, B., “FDA's Pathway for Regulation of FMT: Not So Fraught,” Journal of Law and the Biosciences 2, no. 3 (2015): 742746; Ossorio and Zhou, supra note 67. The controversy is described in Lietzan, supra note 10, at 1254–1257.CrossRefGoogle Scholar
Lietzan, supra note 10, at 1295 (“Access-before-evidence scenarios provide a powerful rejoinder to any argument that, without a federal gatekeeper, competitive market pressures and liability exposure would ensure that new medical treatments are subjected to modern rigorous safety and effectiveness trials. At least for unapproved prescription drugs, compounded drugs, and fecal microbiota, this proposition is manifestly false.”).Google Scholar
U.S. Food and Drug Administration, Guidance for Industry: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies (February 2014), available at <https://www.govinfo.gov/content/pkg/FR-2014-02-26/pdf/FR-2014-02-26.pdf> (last visited September 18, 2019); U.S. Food and Drug Administration, supra note 83.+(last+visited+September+18,+2019);+U.S.+Food+and+Drug+Administration,+supra+note+83.>Google Scholar
U.S. Food and Drug Administration, supra note 83.Google Scholar
Outterson, K., “Regulating Compounding Pharmacies after NECC,” The New England Journal of Medicine 367, no. 21 (2012): 19691972; Centers for Disease Control and Prevention, Multistate Outbreak of Fungal Meningitis and Other Infections (October 30, 2015), available at <https://www.cdc.gov/hai/outbreaks/meningitis.html> (last visited September 18, 2019). In January of 2018, the supervisory pharmacist who oversaw the distribution of contaminated drugs from the compounding pharmacy was convicted of racketeering, conspiracy, mail fraud, and introduction of misbranded drugs into interstate commerce, and sentenced to eight years in prison, two years of supervised release, and forfeiture and restitution to be determined. U.S. Food and Drug Administration, January 31, 2018: New England Compounding Center Pharmacist Sentenced for Role in Nationwide Fungal Meningitis Outbreak (January 31, 2018), available at <https://www.fda.gov/inspections-compliance-enforcement-and-criminal-investigations/criminal-investigations/january-31-2018-new-england-compounding-center-pharmacist-sentenced-role-nationwide-fungal> (last visited September 18, 2019).CrossRefGoogle Scholar
Comments on FDA-2013-D-0811, Guidance for Industry: Enforcement Policy Regarding IND Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium Difficile infection Not Responsive to Standard Therapies, available at <https://www.regulations.gov/docketBrowser?rpp=25&so=DESC‚commentDueDate£25&dct=PS&D=FDA-2013-D-0811> (last visited September 18, 2019).+(last+visited+September+18,+2019).>Google Scholar
OpenBiome, Regulatory, OpenBiome Website, available at <https://www.openbiome.org/regulatory/> (last visited September 18, 2019).+(last+visited+September+18,+2019).>Google Scholar
21 C.F.R. § 314.420(a). A drug master file consists of important regulatory information submitted to the FDA. The holder of the master file can authorize other persons to rely on the master file in support of the other person's IND submission. The holder of the drug master file can keep information in confidence, even withholding the information from persons the holder has authorized to reference the file.Google Scholar
OpenBiome, supra note 54; Rebiotix, supra note 116; Microbiome Therapeutics Innovation Group, Microbiome Therapeutics Innovation Group Position Statement on FMT, available at <https://microbiometig.org/wp-content/uploads/2019/03/MTIG-Position-Statement-FINAL-1.pdf> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
See Khoruts, A., Comment on the Food and Drug Administration (FDA) Notice: Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridium difficile Infection Not Responsive to Standard Therapies: Draft Guidance for Industry (April 25, 2016), Regulations.gov Website, available at <https://www.regulations.gov/document?D=FDA-2013-D-0811-0065> (last visited September 17, 2019). Note that OpenBiome's website claimed the organization had shipped over 43,000 doses of stool as of January, 2019. However, because some people might have received more than one dose and some might not have received the intervention after their provider ordered the stool, one cannot determine how many people have received FMT using stool from OpenBiome. OpenBiome, Our Impact (January, 2019), available at <https://www.openbiome.org/impact> (last visited September 17, 2019). (last visited September 17, 2019).' href=https://scholar.google.com/scholar?q=See+Khoruts,+A.,+Comment+on+the+Food+and+Drug+Administration+(FDA)+Notice:+Enforcement+Policy+Regarding+Investigational+New+Drug+Requirements+for+Use+of+Fecal+Microbiota+for+Transplantation+to+Treat+Clostridium+difficile+Infection+Not+Responsive+to+Standard+Therapies:+Draft+Guidance+for+Industry+(April+25,+2016),+Regulations.gov+Website,+available+at++(last+visited+September+17,+2019).+Note+that+OpenBiome's+website+claimed+the+organization+had+shipped+over+43,000+doses+of+stool+as+of+January,+2019.+However,+because+some+people+might+have+received+more+than+one+dose+and+some+might+not+have+received+the+intervention+after+their+provider+ordered+the+stool,+one+cannot+determine+how+many+people+have+received+FMT+using+stool+from+OpenBiome.+OpenBiome,+Our+Impact+(January,+2019),+available+at++(last+visited+September+17,+2019).>Google Scholar
Kelly, C.R., Fischer, M., Grinspan, A., Allegretti, J.R., “Patients Eligible for Trials of Microbe-Based Therapeutics Do Not Represent the Population with Recurrent Clostridioides difficile Infection,” Clinical Gastroenterology and Hepatology (2019) [epub ahead of print], available at <https://doi.org/10.1016/j.cgh.2019.06.034> (last visited September 17, 2019) (Finding that 17% of rCDI patients who were eligible for a clinical trial testing an oral stool-derived product refused to participate; one reason patients gave was because they might be randomized to a placebo group.).Google Scholar
Hoffmann, et al., supra note 117. One firm with a stool-derived product currently in clinical trials recently stated: “Prospective enrollees have repeatedly told principal investigators that they will forgo involvement in the clinical trial and instead obtain an unapproved product [for FMT] sold by a stool bank over the internet.” Rebiotix, supra note 116, at 2.Google Scholar
McDonald, et al., supra note 53 and accompanying text.Google Scholar
21 C.F.R. § 312.315.Google Scholar
An investigator is “an individual who actually conducts a clinical investigation (i.e., under whose immediate direction the drug is administered or dispensed to a subject). In the event an investigation is conducted by a team of individuals, the investigator is the responsible leader of the team.” 21 C.F.R. § 312.3.Google Scholar
21 C.F.R. § 312.305(a)(3).Google Scholar
A sponsor-investigator is “an individual who both initiates and conducts an investigation, and under whose immediate direction the investigational drug is administered or dispensed.” 21 C.F.R. § 312.3.Google Scholar
Khoruts, supra note 127; Kelly, C.R., Kunde, S.S., and Kho-ruts, A., “Guidance on Preparing an Investigational New Drug Application for Fecal Microbiota Transplantation Studies,” Clinical Gastroenterology and Hepatology 12, no. 2 (2014): 283288. Note that a research-IND is used when there is no commercial sponsor for an FDA-regulated study.CrossRefGoogle Scholar
Id.; Holbien, M.E.B., “Understanding FDA Regulatory Requirements for Investigational New Drug Applications for Sponsor-Investigators,” Journal of Investigative Medicine 57, no. 6 (2009): 688694.CrossRefGoogle Scholar
U.S. Food and Drug Administration, Investigator-Initiated Investigational New Drug (IND) Applications (February 22, 2018), available at <https://www.fda.gov/drugs/investigational-new-drug-ind-application/investigator-initiated-investigational-new-drug-ind-applications> (last visited June 3, 2019).+(last+visited+June+3,+2019).>Google Scholar
21 C.F.R. § 312.3.Google Scholar
21 C.F.R. § 50.3(c).Google Scholar
Kelly, et al., supra note 128.Google Scholar
See, e.g., Kim, E.S., Atlas, J., Ison, G., J and Ersek, L., “Transforming Clinical Trial Eligibility Criteria to Reflect Practical Application” American Society of Clinical Oncology 35 (2016): 8390; see, also, U.S. Food and Drug Administration, Cancer Clinical Trial Eligibility Criteria: Patients with Organ Dysfunction or Prior or Concurrent Malignancies, Draft Guidance for Industry, March 2019, available at <https://www.fda.gov/media/123745/download> (last visited August 3, 2019) (stating that “Unnecessarily restrictive eligibility criteria may slow patient accrual, limit patients' access to clinical trials, and lead to trial results that do not fully represent treatment effects in the patient population that will ultimately use the drug.”).Google Scholar
21 C.F.R. § 312.3.Google Scholar
21 C.F.R. § 312.315(c)(3).Google Scholar
21 C.F.R. § 312.305(c)(5).Google Scholar
Cross ref. to Khoruts, et al., supra note 8.Google Scholar
See Freeman, J., “Collaborative Governance in the Administrative State,” UCLA Law Review 45, no. 1 (1997): 198; Dorf, M.C. and Sabel, C.F., “A Constitution of Democratic Experimentalism,” Columbia Law Review 98, no. 2 (1998): 267473; Lobel, O., “The Renew Deal: The Fall of Regulation and the Rise of Governance in Contemporary Legal Thought,” Minnesota Law Review 89, no. 2 (2004): 342470; Burris, S., Drahos, P., and Shearing, C., “Nodal Governance,” Australasian Journal of Legal Philosophy 30 (2005): 3058; Solomon, J.M., “Law and Governance in the 21st Century Regulatory State,” Texas Law Review 86, no. 4 (2008): 819856.Google Scholar
Lobel, O., “New Governance as Regulatory Governance,” in Levi-Four, D., ed., The Oxford Handbook of Governance (New York: Oxford University Press, 2012): at 6582; Solomon, Id.; Trubek, L.G., “New Governance and Soft Law in Health Care Reform,” Indiana Health Law Review 3, no. 1 (2006): 137169.Google Scholar
U.S. Food and Drug Administration, Guidance for Industry: E9 Statistical Principles for Clinical Trials (September 1998), FDA website, available at <https://www.fda.gov/regulatory-information/search-fda-guidance-documents/e9-statistical-principles-clinical-trials> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
Johnsen, P.H., “Faecal Microbiota Transplantation Versus Placebo for Moderate-to-Severe Irritable Bowel Syndrome: A Double-Blind, Randomized, Placebo-Controlled, Parallel-Group, Single-Centre Trial,” Lancet Gastroenterology & Hepatology 3, no. 1 (2018): 1724; National Institutes of Health, supra note 50.CrossRefGoogle Scholar
Park, et al., supra note 71.Google Scholar
U.S. Food and Drug Administration, Qualifying a Biomarker Through the Biomarker Qualification Program (August 2, 2018), FDA Website, available at <https://www.fda.gov/drugs/cder-biomarker-qualification-program/qualifying-bio-marker-through-biomarker-qualification-program> (last visited September 17, 2019).+(last+visited+September+17,+2019).>Google Scholar
United States v. Regenerative Sciences, LLC, 741 F. 3d 1314 (D.C. Cir. 2014); United States of America v. U.S. Stem Cell Clinic, LLC, et al., United States District Court for the Southern District of Florida, Case No. 0:18-cv-61047-UU, June 3, 2019; see also Grady, D., Can, F.D.A. Act Against Stem Cell Clinic, Judge Rules, The New York Times, June 3, 2019, available at <https://www.nytimes.com/2019/06/03/health/stem-cell-fda-regulate.html> (last visited September 17, 2019).Google Scholar